Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Publication year range
1.
Bioorg Chem ; 117: 105441, 2021 12.
Article in English | MEDLINE | ID: mdl-34695731

ABSTRACT

Under the guidance of bioassay against HSC-LX2, the EtOH extract and the EtOAc fraction of Artemisia capillaris (Yin-Chen) exhibited cytotoxic activity against HSC-LX2 with inhibitory ratios of 39.7% and 68.7% at the concentration of 400.0 µg/mL. Bioassay-guided investigation of Fr. D (the active fraction) yielded 14 new coumaric acid analogues, artemicapillasins A-N (1-14). The structures of the isolates were elucidated by spectroscopic analyses involving UV, IR, MS, 1D and 2D NMR spectra and ECD calculations. Cytotoxic activity against HSC-LX2 cells of these isolates was performed to reveal that 12 compounds demonstrated cytotoxicity with inhibitory ratios more than 50% at 400 µM. The most active artemicapillasin B (2) gave an IC50 value of 24.5 µM, which was about 7 times more toxic than the positive drug silybin (IC50, 162.3 µM). Importantly, artemicapillasin B (2) showed significant inhibition on the deposition of human collagen type I (Col I), human laminin (HL) and human hyaluronic acid (HA) with IC50 values of 11.0, 14.4 and 13.8 µM, which was about 7, 11 and 5 times more active than silybin. Artemicapillasin B (2) as an interesting antihepatic fibrosis candidate is worth in-depth study.


Subject(s)
Artemisia/chemistry , Hepatic Stellate Cells/drug effects , Cell Survival/drug effects , Collagen Type I/antagonists & inhibitors , Collagen Type I/metabolism , Dose-Response Relationship, Drug , Humans , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/metabolism , Laminin/antagonists & inhibitors , Laminin/metabolism , Molecular Structure , Structure-Activity Relationship
2.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502259

ABSTRACT

This study probed the largely unexplored regulation and role of fibronectin in Angiotensin II-stimulated cardiac fibroblasts. Using gene knockdown and overexpression approaches, Western blotting, and promoter pull-down assay, we show that collagen type I-activated Discoidin Domain Receptor 2 (DDR2) mediates Angiotensin II-dependent transcriptional upregulation of fibronectin by Yes-activated Protein in cardiac fibroblasts. Furthermore, siRNA-mediated fibronectin knockdown attenuated Angiotensin II-stimulated expression of collagen type I and anti-apoptotic cIAP2, and enhanced cardiac fibroblast susceptibility to apoptosis. Importantly, an obligate role for fibronectin was observed in Angiotensin II-stimulated expression of AT1R, the Angiotensin II receptor, which would link extracellular matrix (ECM) signaling and Angiotensin II signaling in cardiac fibroblasts. The role of fibronectin in Angiotensin II-stimulated cIAP2, collagen type I, and AT1R expression was mediated by Integrin-ß1-integrin-linked kinase signaling. In vivo, we observed modestly reduced basal levels of AT1R in DDR2-null mouse myocardium, which were associated with the previously reported reduction in myocardial Integrin-ß1 levels. The role of fibronectin, downstream of DDR2, could be a critical determinant of cardiac fibroblast-mediated wound healing following myocardial injury. In summary, our findings suggest a complex mechanism of regulation of cardiac fibroblast function involving two major ECM proteins, collagen type I and fibronectin, and their receptors, DDR2 and Integrin-ß1.


Subject(s)
Discoidin Domain Receptor 2/deficiency , Discoidin Domain Receptor 2/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Integrin beta1/metabolism , Myocardium/metabolism , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II/pharmacology , Animals , Apoptosis/genetics , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Collagen Type I/antagonists & inhibitors , Collagen Type I/metabolism , Discoidin Domain Receptor 2/genetics , Fibroblasts/drug effects , Fibronectins/genetics , Gene Knockdown Techniques , Gene Silencing , Heart/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Mice , Mice, Knockout , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Signal Transduction , YAP-Signaling Proteins
3.
Molecules ; 25(4)2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32054011

ABSTRACT

Twenty 9O-substituted palmatine derivatives were prepared and tested for their biological effect against collagen α1 (I) (COL1A1) promotor in human hepatic stellate LX-2 cells. The structure-activity relationship (SAR) indicated that the introduction of a benzyl motif on the 9O atom was favorable for activity. Among them, compound 6c provided the highest inhibitory effect against COL1A1 with an IC50 value of 3.98 µM, and it also dose-dependently inhibited the expression of fibrogenic COL1A1, α-soomth muscle actin (α-SMA), matrix metalloprotein 2 (MMP2) in both mRNA and protein levels, indicating extensive inhibitory activity against fibrogenesis. A further primary mechanism study indicated that it might repress the hepatic fibrogenesis via inhibiting both canonical transforming growth factor-beta 1 (TGF-ß1)/Smads and non-canonical janus-activated kinase 1 (JAK1)/singal transducer and activator of transcription 3 (STAT3) signaling pathways. Additionally, 6c owned a high safety profile with the LD50 value of over 1000 mg·kg-1 in mice. These results identified palmatine derivatives as a novel class of anti-fibrogenic agents, and provided powerful information for further structure optimization.


Subject(s)
Berberine Alkaloids/chemistry , Berberine Alkaloids/pharmacology , Collagen Type I/antagonists & inhibitors , Janus Kinase 1/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Dose-Response Relationship, Drug , Gene Expression , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Models, Biological , Molecular Structure , Structure-Activity Relationship
4.
Am J Physiol Renal Physiol ; 317(6): F1430-F1438, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31545926

ABSTRACT

Renal fibrosis is a key pathological phenomenon of chronic kidney disease (CKD) contributing to the progressive loss of renal function. UK383,367 is a procollagen C proteinase inhibitor that has been selected as a candidate for dermal antiscarring agents, whereas its role in renal fibrosis is unclear. In the present study, UK383,367 was applied to a CKD mouse model of unilateral ureteral obstruction (UUO) and cell lines of renal tubular epithelial cells (mouse proximal tubular cells) and renal fibroblast cells (NRK-49F cells) challenged by transforming growth factor-ß1. In vivo, bone morphogenetic protein 1, the target of UK383,367, was significantly enhanced in UUO mouse kidneys and renal biopsies from patients with CKD. Strikingly, UK383,367 administration ameliorated tubulointerstitial fibrosis as shown by Masson's trichrome staining in line with the blocked expression of collagen type I/III, fibronectin, and α-smooth muscle actin in the kidneys from UUO mice. Similarly, the enhanced inflammatory factors in obstructed kidneys were also blunted. In vitro, UK383,367 pretreatment inhibited the induction of collagen type I/III, fibronectin, and α-smooth muscle actin in both mouse proximal tubular cells and NRK-49F cells treated with transforming growth factor-ß1. Taken together, these findings indicate that the bone morphogenetic protein 1 inhibitor UK383,367 could serve as a potential drug in antagonizing CKD renal fibrosis by acting on the maturation and deposition of collagen and the subsequent profibrotic response and inflammation.


Subject(s)
Bone Morphogenetic Protein 1/antagonists & inhibitors , Oxadiazoles/therapeutic use , Renal Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Animals , Cell Line , Child , Child, Preschool , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Collagen Type III/antagonists & inhibitors , Collagen Type III/biosynthesis , Female , Fibronectins/antagonists & inhibitors , Fibronectins/biosynthesis , Fibrosis/drug therapy , Humans , Inflammation/pathology , Inflammation/prevention & control , Kidney/pathology , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Ureteral Obstruction/complications
5.
Langmuir ; 35(4): 980-989, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30621400

ABSTRACT

Type I collagen is a fibrous protein, which is highly biocompatible and biodegradable and exhibits low immunogenicity with its unique feature of undergoing a spontaneous self-assembly process. However, the excessive accumulation of collagen may lead to a condition known as fibrosis in vertebrates. Recently, saturated fatty acids have gained much attention as biomedical and therapeutic agents. Therefore, drawing inspiration from the biological and structural tunability of these fatty acids, this work aims to inhibit the self-assembly of type I collagen using (±)-α-lipoic acid (ALA). Reconstituted collagen and its blends with (±)-ALA under physiological conditions were subjected to fibril growth kinetics measurements, which exhibited the decrease in the rate of fibrillogenesis ( t1/2) with an increase in the concentration of ALA. Variations in the viscoelasticity of collagen and ALA blend with respect to rate and frequency showed significant changes. Further, the frequency shifts of different functional groups via FT-IR (ATR) and the morphological changes associated with fibril inhibition were visualized using a cryoscanning electron microscope. Molecular dynamics simulation of the collagen-like peptide with the (±)-ALA molecule at different molar ratios proved that (±)-ALA had a strong potential to bind at various sites of collagen mediated by conventional secondary or noncovalent forces. Thus, the protein-small molecule interaction dominates the forces prevailing between protein-protein binding, leading to the inhibition of the self-assembly process. Such inhibitory effects by a fatty acid may unfold newer avenues for development of targeted and sustainable drug delivery systems for fibrotic diseases.


Subject(s)
Collagen Type I/antagonists & inhibitors , Thioctic Acid/pharmacology , Animals , Collagen Type I/chemistry , Collagen Type I/isolation & purification , Molecular Dynamics Simulation , Rats , Rats, Wistar
6.
Int J Med Sci ; 16(1): 23-32, 2019.
Article in English | MEDLINE | ID: mdl-30662325

ABSTRACT

Bisphosphonates (BPs) represent the first-line treatment for a wide array of bone disorders. Despite their well-known action on osteoclasts, the effects they induce on osteoblasts are still unclear. In order to shed light on this aspect we evaluated the impact of two nitrogen containing bisphosphonates, Alendronate (ALN) and Zoledronate (ZOL), on human primary pre-osteoblasts. At first, we showed an inhibitory effect on cell viability and alkaline phosphatase activity starting from µM concentrations of both drugs. In addition, an inhibitory trend on mineralized nodules deposition was observed. Then low doses of both ALN and ZOL rapidly increased the release of the pro-inflammatory mediators TNFα and IL-1ß, while increased DKK-1 and Sclerostin, both inhibitors of osteoblastogenesis. Finally, ALN and 10-7M ZOL decreased the expression of type I Collagen and Osteopontin, while both drugs slightly stimulated SPARC production. With these results, we would like to suggest a direct inhibitory action on bone-forming cells by nitrogen containing bisphosphonates.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Osteoblasts/metabolism , Zoledronic Acid/pharmacology , Alendronate/therapeutic use , Alkaline Phosphatase/antagonists & inhibitors , Biomarkers/metabolism , Bone Density Conservation Agents/therapeutic use , Bone Diseases/drug therapy , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen Type I/antagonists & inhibitors , Cytokines/metabolism , Humans , Osteoblasts/cytology , Osteoblasts/drug effects , Osteopontin/antagonists & inhibitors , Zoledronic Acid/therapeutic use
7.
Molecules ; 24(20)2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31627430

ABSTRACT

Twenty new 12N-substituted matrinol derivatives were synthesized and evaluated for their inhibitory effects on collagen α1 (I) (COL1A1) promotor in human hepatic stellate LX-2 cells. The structure-activity relationship (SAR) revealed that introducing a 12N-benzeneaminoacylmethyl substitution might significantly enhance the activity. Compound 8a exhibited the highest inhibitory potency against COL1A1, and its inhibition activity against COL1A1 was further confirmed on both the mRNA and protein levels. It also effectively inhibited the expression of α smooth muscle actin (α-SMA), fibronectin and transforming growth factor ß1 (TGFß1), indicating an extensive inhibitory effect on the expression of fibrogenic genes. The primary mechanism study indicated that it might take action via the Integrin/FAK/PI3K/Akt signaling pathway. The results provided powerful information for further structure optimization, and compound 8a was selected as a novel anti-fibrogenic lead for further investigation.


Subject(s)
Collagen Type I/genetics , Hepatic Stellate Cells/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Signal Transduction/drug effects , Thiazoles/pharmacology , Cell Line , Collagen Type I/antagonists & inhibitors , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibrosis/prevention & control , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Humans , Integrin alphaV/genetics , Integrin alphaV/metabolism , Models, Biological , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Structure-Activity Relationship , Thiazoles/chemical synthesis
8.
J Cell Mol Med ; 22(3): 2018-2022, 2018 03.
Article in English | MEDLINE | ID: mdl-29278308

ABSTRACT

Penile fibrosis caused by ischemic priapism (IP) adversely affects patients' erectile function. We explored the role of lysyl oxidase (LOX) in rat and human penes after ischemic priapism (IP) to verify the effects of anti-LOX in relieving penile fibrosis and preventing erectile dysfunction caused by IP in rats. Seventy-two rats were randomly divided into six groups: control group, control + ß-aminopropionitrile (BAPN) group, 9 hrs group, 9 hrs + BAPN group, 24 hrs group, and 24 hrs + BAPN group. ß-aminopropionitrile (BAPN), a specific inhibitor of LOX, was administered in the drinking water. At 1 week and 4 weeks, half of the rats in each group were randomly selected for the experiment. Compared to the control group, the erectile function of IP rats was significantly decreased while the expression of LOX in the corpus cavernosum was significantly up-regulated in both 9 and 24 hrs group. Proliferated fibroblasts, decreased corpus cavernosum smooth muscle cells/collagen ratios, destroyed endothelial continuity, deposited abnormal collagen and disorganized fibers were observed in IP rats. The relative content of collage I and III was not obviously different among the groups. ß-aminopropionitrile (BAPN) could effectively improve the structure and erectile function of the penis, and enhance recovery. The data in this study suggests that LOX may play an important role in the fibrosis of corpus cavernosum after IP and anti-LOX may be a novel target for patients suffering with IP.


Subject(s)
Aminopropionitrile/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Ischemia/drug therapy , Priapism/prevention & control , Animals , Cell Proliferation/drug effects , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/antagonists & inhibitors , Collagen Type III/genetics , Collagen Type III/metabolism , Disease Models, Animal , Drinking Water/administration & dosage , Fibroblasts/enzymology , Fibroblasts/pathology , Fibrosis/prevention & control , Gene Expression , Humans , Ischemia/enzymology , Ischemia/genetics , Ischemia/physiopathology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Penile Erection/physiology , Penis/enzymology , Penis/physiopathology , Priapism/enzymology , Priapism/genetics , Priapism/physiopathology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Rats , Rats, Sprague-Dawley
9.
Biochem Cell Biol ; 96(1): 19-25, 2018 02.
Article in English | MEDLINE | ID: mdl-28482162

ABSTRACT

Gastric cancer (GC) is one of the most lethal cancers worldwide. In this study, we aimed to explore the role of miR-129-5p, a newly identified miR-129 member, in GC cells as well as the potential mechanism of action. The results of reverse transcription - qualitative polymerase chain reaction (RT-qPCR) and Western Blot showed that miR-129 was downregulated in GC cells compared with normal ones. Using MTT, colony formation, wound healing assay, and a Transwell assay, we evaluated the proliferation, migration, and invasion abilities of transfected cells, and confirmed miR-129-5p as a tumor suppressor in GC. After a microarray analysis comparing different gene expressions in miR-129-5p transfected SGC-7901 cells, COL1A1 was selected for biggest fold-change and potential target of miR-129-5p predicted by TargetScan. Measured by RT-qPCR and Western blot, COL1A1 turned out to be upregulated in GC tissues and cells. We further confirmed the targeting relationship between miR-129-5p and COL1A1 by dual luciferase assay. By manipulating the expression of COL1A1 in SGC-7901 cells, cell proliferation, migration, and invasion were examined and the tumor-promoting function of COL1A1 was validated. Moreover, co-transfection of miR-129-5p mimics and COL1A1 attenuated the tumor-promoting effects induced by a single-transfection of COL1A1, and miR-129-5p inhibitor counteracted the tumor-suppressing effects of COL1A1 siRNA. Collectively, the data demonstrate the important functions of the miR-129-5p-COL1A1 axis in GC: miR-129-5p suppresses GC cell proliferation, migration, and invasion, by selectively inhibiting COL1A1. This study provides new therapeutic targets for the clinical treatment of GC.


Subject(s)
Collagen Type I/antagonists & inhibitors , MicroRNAs/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Female , Humans , Male , MicroRNAs/genetics , Stomach Neoplasms/metabolism
10.
Toxicol Appl Pharmacol ; 355: 127-137, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30008374

ABSTRACT

Fibrosis is a pathophysiological state characterized by the excessive formation/deposition of fibrous extracellular matrix. Transforming growth factor-beta (TGF-ß) is a central profibrotic mediator, and targeting TGF-ß is a promising strategy in the development of drugs for the treatment of fibrosis. Therefore, the effect of LY2109761, a small molecule inhibitor against TGF-ß with targets beyond TGF-ß signaling, on fibrogenesis was elucidated in vitro (HepG2 cells and LX-2 cells) and ex vivo (human and rat precision-cut liver slices). Our results displayed an anti-fibrotic effect of LY2109761, as it markedly down-regulated gene and protein expression of collagen type 1, as well as gene expression of the inhibitor of metalloproteinases 1. This effect on fibrosis markers was partially mediated by targeting TGF-ß signaling, seeing that LY2109761 inhibited TGF-ß1 gene expression and SMAD2 protein phosphorylation. Interestingly, particularly at a high concentration, LY2109761 decreased SMAD1 protein phosphorylation and gene expression of the inhibitor of DNA binding 1, which appeared to be TGF-ß-independent effects. In conclusion, LY2109761 exhibited preclinical anti-fibrotic effects via both TGF-ß-dependent and -independent pathways. These results illustrate that small molecule inhibitors directed against TGF-ß could possibly influence numerous signaling pathways and thereby mitigate fibrogenesis.


Subject(s)
Fibrosis/drug therapy , Pyrazoles/pharmacology , Pyrroles/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Cell Line , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Down-Regulation , Gene Expression/drug effects , Humans , In Vitro Techniques , Liver/drug effects , Liver/metabolism , Male , Phosphorylation , Rats , Rats, Wistar , Smad1 Protein/antagonists & inhibitors , Smad2 Protein/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-2/antagonists & inhibitors
11.
Toxicol Appl Pharmacol ; 339: 172-180, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29253500

ABSTRACT

Carnosic acid (CA), a major bioactive component in rosemary extract, has many biological and pharmaceutical activities. Smad3 acetylation can regulate the transcription of type I α2 collagen (COL1A2), which is the major component of the extracellular matrix (ECM). The aim of the current study was to evaluate whether CA inhibits COL1A2 transcription via the reduction of Smad3 acetylation against liver fibrosis. The results showed that CA treatment significantly suppressed COL1A2 transcription and markedly decreased the deposition of ECM induced by dimethylamine (DMN) in rats. Importantly, the suppression of COL1A2 transcription following CA treatment depended on the reduction of Smad3 acetylation via the activation of Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide+ (NAD+)-dependent deacetylase. SIRT1 siRNA increased the acetylation of Smad3 and blocked CA-down-regulated Smad3 deacetylation. Notably, CA-mediated AMP-activated protein kinase-α1 (AMPKα1) activation not only increased AMPKα1 phosphorylation but also increased SIRT1 expression, thus leading to a significant reduction in Smad3 acetylation. Furthermore, CA-mediated SIRT1 activation was inhibited by AMPKα1 siRNA. Collectively, CA can inhibit the transcription of COL1A2 through SIRT1-mediated Smad3 deacetylation, and the activation of SIRT1 by CA involves the AMPKα1/SIRT1 pathway in liver fibrosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Abietanes/pharmacology , Collagen Type I/metabolism , Sirtuin 1/metabolism , Smad3 Protein/metabolism , Transcription, Genetic/physiology , Acetylation/drug effects , Animals , Antioxidants/pharmacology , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Liver Cirrhosis/metabolism , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects
12.
Acta Pharmacol Sin ; 39(2): 195-204, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28905939

ABSTRACT

Cardiac fibrosis is considered the initial change of diabetic cardiomyopathy (DCM). We have shown that curcumin alleviates collagen deposition in DCM, but the mechanism remains unknown. In this study we sought to investigate the effects of curcumin on cardiac fibrosis in vivo and in vitro and to elucidate the underlying mechanisms. Experimental diabetes was induced in rats by injection of low-dose streptozotocin (STZ) combined with high energy diet. The rats were orally treated with curcumin (300 mg·kg-1·d-1) for 16 weeks. Curcumin administration significantly suppressed the deposition of type I and type III collagens in the heart tissues of diabetic rats, accompanied by markedly reduced TGF-ß1 production, suppressed TßR II levels and Smad2/3 phosphorylation, and increased Smad7 expression. Similar effects were observed in human cardiac fibroblasts exposed to high glucose (HG, 30 mmol/L) or exogenous TGF-ß1 (5 ng/mL). Furthermore, TGF-ß1 or HG treatment significantly increased the phosphorylation levels of AMPK and p38 MAPK in the fibroblasts. Application of curcumin (25 µmol/L) inhibited TGF-ß1- or HG-induced AMPK/p38 MAPK activation and suppressed collagen synthesis in the fibroblasts. These effects were similar to those of the AMPK inhibitor compound C (10 µmol/L) but opposite to the effects of the AMPK activator metformin (2 mmol/L) in the fibroblasts. Our results demonstrate that curcumin suppresses diabetes-associated collagen synthesis in rat myocardium not only by inhibiting TGF-ß1 production and canonical Smad signaling but also by blocking the non-canonical AMPK/p38 MAPK pathway.


Subject(s)
Collagen Type III/antagonists & inhibitors , Collagen Type I/antagonists & inhibitors , Curcumin/pharmacology , Diabetes Mellitus, Experimental/metabolism , Myocardium/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Diabetic Cardiomyopathies/metabolism , Disease Models, Animal , Fibroblasts/drug effects , Fibrosis/prevention & control , Glucose/metabolism , Humans , Male , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Cell Mol Med ; 21(12): 3506-3514, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28767184

ABSTRACT

Acute kidney disease (AKI) leads to increased risk of progression to chronic kidney disease (CKD). Antithrombin III (ATIII) is a potent anticoagulant with anti-inflammatory properties, and we previously reported that insufficiencies of ATIII exacerbated renal ischaemia-reperfusion injury (IRI) in rats. In this study, we examined the characteristic of AKI-CKD transition in rats with two distinct AKI models. Based on our observation, left IRI plus right nephrectomy (NX-IRI) was used to determine whether ATIII had therapeutic effects in preventing CKD progression after AKI. It was observed that NX-IRI resulted in significant functional and histological damage at 5 weeks after NX-IRI compared with sham rats, which was mitigated by ATIII administration. Besides, we noticed that ATIII administration significantly reduced NX-IRI-induced interstitial fibrosis. Consistently, renal expression of collagen-1, α-smooth muscle actin and fibronectin were substantial diminished in ATIII-administered rats compared with un-treated NX-IRI rats. Furthermore, the beneficial effects of ATIII were accompanied with decreased M1-like macrophage recruitment and down-regulation of M1-like macrophage-dependent pro-inflammatory cytokines such as tumour necrosis factor α, inducible nitric oxide synthase and interleukin-1ß, indicating that ATIII prevented AKI-CKD transition via inhibiting inflammation. Overall, ATIII shows potential as a therapeutic strategy for the prevention of CKD progression after AKI.


Subject(s)
Acute Kidney Injury/prevention & control , Antithrombin III/pharmacology , Gene Expression Regulation/drug effects , Kidney/drug effects , Reperfusion Injury/prevention & control , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Disease Models, Animal , Disease Progression , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kidney/metabolism , Kidney/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Nephrectomy , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Nitric Oxide ; 70: 31-41, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28757441

ABSTRACT

OBJECTIVE: Inducible nitric oxide synthase (iNOS) aggravates and endothelial nitric oxide synthase (eNOS) ameliorates fibrosis in the lung. Our previous study demonstrated that aminoguanidine (AG), a preferred iNOS inhibitor, prevents bleomycin-induced injury and fibrosis in the lung. The diethylenetriamine nitric oxide adduct (DETA/NO) is a slow-release NO donor. Here, to clarify the exact role of the nitric oxide (NO) molecule in the pathogenesis of pulmonary fibrosis in vivo, we observed the effects of inhalation of aerosolized DETA/NO on fibrosis in the lungs of bleomycin-exposed rats with AG treatment, including the effects on the myofibroblast number, collagen deposition, peroxynitrite anion (ONOO-) formation, and injury in the lung. DESIGN AND METHODS: Rats received a single intratracheal instillation of bleomycin or normal saline (NS) on day 0, followed by a daily intraperitoneal injection of AG or NS from day 1 to day 13. Each group was additionally given a daily inhalation of DETA/NO or placebo from day 1 to day 13. On day 14, half of the rats in each group was euthanized, and plasma nitrite and nitrate (NOx), myofibroblasts, type I collagen, ONOO- and injury in the lung were estimated by the Griess reaction, western blotting, immunohistochemical staining, sirius red staining, and hematoxylin and eosin (HE) staining, respectively. On day 28, the other half of the rats in each group was euthanized, and the total collagen of the lung was evaluated by hydroxyproline assay. RESULTS: ① At the day 14 time point, AG reduced the plasma NOx level in bleomycin rats, while this drug had no significant effect on sham rats. Inhalation of aerosolized DETA/NO increased the plasma NOx level of bleomycin + AG rats, sham rats and sham + AG rats. However, due to large areas of airspace obliteration in the lungs of bleomycin rats, DETA/NO inhalation had no significant effect on the plasma NOx level in these rats. ② At the day 14 time point, AG reduced ONOO- formation (marked by nitrotyrosine, NT), injury, myofibroblast number, and type I collagen deposition in the lungs of bleomycin rats, while this drug had no significant impact on the above parameters in the lungs of sham rats. Interestingly, DETA/NO inhalation enhanced the preventive effects afforded by AG on myofibroblast number and type I collagen deposition, but had no significant impact on ONOO- and injury in lung. ③ At the day 28 time point, because rats were not exposed to DETA/NO after day 13, there was no significant difference of the plasma NOx level in sham rats, sham + AG rats, bleomycin rats, and bleomycin + AG rats between DETA/NO inhalation and placebo inhalation. Interestingly, rats administered both DETA/NO and AG still showed a reduction in total collagen of the entire lung compared to rats administered AG alone at this time point. CONCLUSIONS: Exogenous NO enhances the prophylactic effect afforded by AG on the myofibroblast number and collagen deposition in the lungs of bleomycin-treated rats in vivo. These results suggest that NO has a direct antifibrotic effect in lungs, except for the formation of ONOO- in the development of pulmonary fibrosis in vivo.


Subject(s)
Guanidines/therapeutic use , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pulmonary Fibrosis/prevention & control , Triazenes/therapeutic use , Animals , Bleomycin , Collagen Type I/antagonists & inhibitors , Drug Synergism , Guanidines/administration & dosage , Male , Myofibroblasts/drug effects , Nitrates/blood , Nitric Oxide/physiology , Nitric Oxide Donors/administration & dosage , Nitrites/blood , Pulmonary Fibrosis/chemically induced , Rats, Sprague-Dawley , Triazenes/administration & dosage
15.
J Mol Cell Cardiol ; 90: 59-69, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26674152

ABSTRACT

Delineation of mechanisms underlying the regulation of fibrosis-related genes in the heart is an important clinical goal as cardiac fibrosis is a major cause of myocardial dysfunction. This study probed the regulation of Discoidin Domain Receptor 2 (DDR2) gene expression and the regulatory links between Angiotensin II, DDR2 and collagen in Angiotensin II-stimulated cardiac fibroblasts. Real-time PCR and western blot analyses showed that Angiotensin II enhances DDR2 mRNA and protein expression in rat cardiac fibroblasts via NADPH oxidase-dependent reactive oxygen species induction. NF-κB activation, demonstrated by gel shift assay, abolition of DDR2 expression upon NF-κB inhibition, and luciferase and chromatin immunoprecipitation assays confirmed transcriptional control of DDR2 by NF-κB in Angiotensin II-treated cells. Inhibitors of Phospholipase C and Protein kinase C prevented Angiotensin II-dependent p38 MAPK phosphorylation that in turn blocked NF-κB activation. Angiotensin II also enhanced collagen gene expression. Importantly, the stimulatory effects of Angiotensin II on DDR2 and collagen were inter-dependent as siRNA-mediated silencing of one abolished the other. Angiotensin II promoted ERK1/2 phosphorylation whose inhibition attenuated Angiotensin II-stimulation of collagen but not DDR2. Furthermore, DDR2 knockdown prevented Angiotensin II-induced ERK1/2 phosphorylation, indicating that DDR2-dependent ERK1/2 activation enhances collagen expression in cells exposed to Angiotensin II. DDR2 knockdown was also associated with compromised wound healing response to Angiotensin II. To conclude, Angiotensin II promotes NF-κB activation that up-regulates DDR2 transcription. A reciprocal regulatory relationship between DDR2 and collagen, involving cross-talk between the GPCR and RTK pathways, is central to Angiotensin II-induced increase in collagen expression in cardiac fibroblasts.


Subject(s)
Angiotensin II/pharmacology , Collagen Type I/genetics , Fibroblasts/drug effects , Myocardium/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Angiotensin II/genetics , Angiotensin II/metabolism , Animals , Collagen Type I/antagonists & inhibitors , Collagen Type I/metabolism , Discoidin Domain Receptors , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocardium/cytology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Primary Cell Culture , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/antagonists & inhibitors , Receptors, Mitogen/metabolism , Signal Transduction , Transcription, Genetic , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
IUBMB Life ; 68(7): 589-96, 2016 07.
Article in English | MEDLINE | ID: mdl-27278959

ABSTRACT

Activation of hepatic stellate cells (HSCs) is characterized by expression of extracellular matrix and loss of adipogenic phenotype during liver fibrogenesis. Emerging evidence suggests that HSCs adopt aerobic glycolysis during activation. The present work aimed at investigating whether the anti-fibrogenic effects of curcumin was associated with interfering with glycolysis in HSCs. Primary rat HSCs were cultured in vitro. We demonstrated that inhibition of glycolysis by 2-deoxyglucose or galloflavin reduced the expression of α-smooth muscle actin (α-SMA) and α1(I)procollagen at both mRNA and protein levels, and increased the intracellular lipid contents and upregulated the gene and protein expression of adipogenic transcription factors C/EBPα and PPAR-γ in HSCs. Curcumin at 20 µM produced similar effects. Moreover, curcumin decreased the expression of hexokinase (HK), phosphofructokinase-2 (PFK2), and glucose transporter 4 (glut4), three key glycolytic parameters, at both mRNA and protein levels. Curcumin also reduced lactate production concentration-dependently in HSCs. Furthermore, curcumin increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK), but AMPK inhibitor BML-275 significantly abolished the curcumin downregulation of HK, PFK2, and glut4. In addition, curcumin inhibition of α-SMA and α1(I)procollagen was rescued by BML-275, and curcumin upregulation of C/EBPα and PPAR-γ was abrogated by BML-275. These results collectively indicated that curcumin inhibited glycolysis in an AMPK activation-dependent manner in HSCs. We revealed a novel mechanism for curcumin suppression of HSC activation implicated in antifibrotic therapy. © 2016 IUBMB Life, 68(7):589-596, 2016.


Subject(s)
AMP-Activated Protein Kinases/genetics , Curcumin/administration & dosage , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/drug therapy , Liver/metabolism , Actins/antagonists & inhibitors , Animals , Collagen Type I/antagonists & inhibitors , Collagen Type I, alpha 1 Chain , Deoxyglucose/biosynthesis , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/biosynthesis , Glycolysis/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Hexokinase/biosynthesis , Humans , Isocoumarins/administration & dosage , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Phosphofructokinase-2/biosynthesis , Rats , Rats, Sprague-Dawley
17.
Can J Physiol Pharmacol ; 94(5): 498-507, 2016 May.
Article in English | MEDLINE | ID: mdl-26854595

ABSTRACT

Hepatic stellate cells (HSCs) activation represents an essential event during alcoholic liver fibrosis (ALF). Previous studies have demonstrated that the rat HSCs could be significantly activated after exposure to 200 µmol/L acetaldehyde for 48 h, and the cAMP/PKA signaling pathways were also dramatically upregulated in activated HSCs isolated from alcoholic fibrotic rat liver. Exchange protein activated by cAMP (EPAC) is a family of guanine nucleotide exchange factors (GEFs) for the small Ras-like GTPases Rap, and is being considered as a vital mediator of cAMP signaling in parallel with the principal cAMP target protein kinase A (PKA). Our data showed that both cAMP/PKA and cAMP/EPAC signaling pathways were involved in acetaldehyde-induced HSCs. Acetaldehyde could reduce the expression of EPAC1 while enhancing the expression of EPAC2. The cAMP analog Me-cAMP, which stimulates the EPAC/Rap1 pathway, could significantly decrease the proliferation and collagen synthesis of acetaldehyde-induced HSCs. Furthermore, depletion of EPAC2, but not EPAC1, prevented the activation of HSC measured as the production of α-SMA and collagen type I and III, indicating that EPAC1 appears to have protective effects on acetaldehyde-induced HSCs. Curiously, activation of PKA or EPAC perhaps has opposite effects on the synthesis of collagen and α-SMA: EPAC activation by Me-cAMP increased the levels of GTP-bound (activated) Rap1 while PKA activation by Phe-cAMP had no significant effects on such binding. These results suggested that EPAC activation could inhibit the activation and proliferation of acetaldehyde-induced HSCs via Rap1.


Subject(s)
Guanine Nucleotide Exchange Factors/agonists , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Alcoholic/metabolism , rap1 GTP-Binding Proteins/agonists , Acetaldehyde/antagonists & inhibitors , Acetaldehyde/toxicity , Actins/agonists , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Collagen Type I/agonists , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/agonists , Collagen Type II/antagonists & inhibitors , Collagen Type II/genetics , Collagen Type II/metabolism , Cyclic AMP/agonists , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Alcoholic/prevention & control , RNA Interference , Rats , Second Messenger Systems/drug effects , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
18.
Wound Repair Regen ; 22(1): 125-33, 2014.
Article in English | MEDLINE | ID: mdl-24471776

ABSTRACT

Simvastatin, a 3-hydroxy-3-methylglutaryl coenzyme-A reductase inhibitor, is used to reduce cholesterol levels. Accumulating evidence has revealed the immunomodulatory and anti-inflammatory effects of simvastatin that prevent cardiovascular diseases. In addition, the beneficial effects of statins on fibrosis of various organs have been reported. However, the functional effect of statins on dermal fibrosis of keloids has not yet been explored. The objective of this study was to determine whether simvastatin could affect dermal fibrosis associated with keloids. We examined the effect of simvastatin on transforming growth factor (TGF)-ß1-induced production of type I collagen, connective tissue growth factor (CTGF or CCN2), and α-smooth muscle actin (α-SMA). Keloid fibroblasts were cultured and exposed to different concentrations of simvastatin in the presence of TGF-ß1, and the effects of simvastatin on TGF-ß1-induced collagen and CTGF production in keloid fibroblasts were determined. The type I collagen, CTGF, and α-SMA expression levels and the Smad2 and Smad3 phosphorylation levels were assessed by Western blotting. The effect of simvastatin on cell viability was evaluated by assessing the colorimetric conversion of 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide. Simvastatin suppressed TGF-ß1-induced type I collagen, CTGF, and α-SMA production in a concentration-dependent manner. The TGF-ß1-induced Smad2 and Smad3 phosphorylation levels were abrogated by simvastatin pretreatment. The inhibition of type I collagen, CTGF, and α-SMA expression by simvastatin was reversed by geranylgeranyl pyrophosphate, suggesting that the simvastatin-induced cellular responses were due to inhibition of small GTPase Rho involvement. A RhoA activation assay showed that preincubation with simvastatin significantly blocked TGF-ß1-induced RhoA activation. The Rho-associated coiled kinase inhibitor Y27632 abrogated TGF-ß1-induced production of type I collagen, CTGF, and α-SMA. However, Y27632 had no significant effect on TGF-ß1-induced phosphorylation of Smad2 and Smad3. In conclusion, the present study suggests that simvastatin is an effective inhibitor of TGF-ß1-induced type I collagen, CTGF, and α-SMA production in keloid fibroblasts.


Subject(s)
Actins/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Collagen Type I/antagonists & inhibitors , Connective Tissue Growth Factor/antagonists & inhibitors , Keloid/metabolism , Simvastatin/pharmacology , Transforming Growth Factor beta1/metabolism , Wound Healing , Actins/metabolism , Amides/pharmacology , Anti-Inflammatory Agents/administration & dosage , Blotting, Western , Cells, Cultured , Collagen Type I/metabolism , Connective Tissue Growth Factor/metabolism , Dose-Response Relationship, Drug , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/prevention & control , Humans , Keloid/pathology , Male , Molecular Sequence Data , Phosphorylation/drug effects , Pyridines/pharmacology , Simvastatin/administration & dosage , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/drug effects , Wound Healing/drug effects , rho-Associated Kinases/metabolism
19.
Genet Mol Res ; 13(3): 6398-410, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25158258

ABSTRACT

This study was designed to investigate the effect of different concentrations of rifampicin on osteogenic differentiation and proliferation of mesenchymal stem cells (MSCs) in human bone marrow. Rifampicin treatment at 0, 4, 8, 16, 32, 64, and 128 mg/mL was applied throughout the whole process, from stromal cells purified from human bone marrow to differentiated bone cells. The effect of rifampicin on MSC proliferation was determined using the MTT assay. The effect of rifampicin on the expressions of type I collagen (COL1A1), osteopontin/bone Gla protein (OPN/BGP), and alkaline phosphatase (ALP) in human osteoblast cells were determined by real-time polymerase chain reaction, and the expressions of COL1A1, OPN/BGP, and the runt-related transcription factor (RUNX2) were determined by Western blot. Results showed that the proliferation of MSCs was significantly inhibited when the rifampicin concentration exceeded 32 mg/mL. In addition, increased rifampicin concentrations inhibited the formation of calcium nodules, OPN/BGP, and COL1A1 in osteoblasts after 28 days of induction. The RNA expressions of OPN/BGP, COL1A1, and ALP were significantly downregulated compared to those of the control group in osteoblasts after induction. The protein expressions of RUNX2, COL1A1, and OPN/BGP were also significantly downregulated compared to those of the control group after induction. In conclusion, rifampicin at exorbitant concentration exerts adverse effects on the proliferation of MSCs in human bone marrow and the differentiation of osteoblasts.


Subject(s)
Antitubercular Agents/pharmacology , Bone Marrow Cells/drug effects , Gene Expression/drug effects , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Rifampin/pharmacology , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dose-Response Relationship, Drug , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteopontin/antagonists & inhibitors , Osteopontin/genetics , Osteopontin/metabolism , Primary Cell Culture
20.
Molecules ; 19(5): 6407-14, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24853614

ABSTRACT

Ultraviolet (UV) exposure is well-known to induce premature aging, which is mediated by matrix metalloproteinase-1 (MMP-1) activity. A 9-mer peptide, CopA3 (CopA3) was synthesized from a natural peptide, coprisin, which is isolated from the dung beetle Copris tripartitus. As part of our continuing search for novel bioactive natural products, CopA3 was investigated for its in vitro anti-skin photoaging activity. UV-induced inhibition of type-I procollagen and induction of MMP-1 were partially prevented in human skin fibroblasts by CopA3 peptide in a dose-dependent manner. At a concentration of 25 µM, CopA3 nearly completely inhibited MMP-1 expression. These results suggest that CopA3, an insect peptide, is a potential candidate for the prevention and treatment of skin aging.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Collagen Type I/antagonists & inhibitors , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Insect Proteins/pharmacology , Matrix Metalloproteinase 1/metabolism , Cells, Cultured , Collagen Type I/radiation effects , Dose-Response Relationship, Drug , Matrix Metalloproteinase 1/genetics , Skin/cytology , Skin/metabolism , Skin/radiation effects , Skin Aging/drug effects , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL