Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 292
Filter
1.
Int J Cosmet Sci ; 43(5): 619-626, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34403541

ABSTRACT

INTRODUCTION: We report on the in vitro and ex vivo effects of chiral (R)-10-hydroxystearic acid (10-HSA) compared with other mono-hydroxystearic acid regioisomers and stearic acid (SA) together with its benefit when combined with retinol. METHODS: Following treatment with hydroxystearic acids peroxisomal proliferator-activated receptor alpha (PPARα) activity was determined in a luciferase reporter gene assay, collagen type I was assessed in primary human dermal fibroblasts by immunohistochemistry, modification of the intracellular fibroblast collagen proteome was studied by mass-spectrometry-based proteomics and collagen type III was assessed by immunohistochemistry on human ex vivo skin. RESULTS: 10-HSA was the most effective PPARα agonist (15.7× induction; p < 0.001), followed by 9-HSA (10.1× induction) and then 12-HSA (4.9× induction) with 17-HSA (1.7× induction) being similar to the effects of stearic acid (1.8× induction). Collagen type I levels were increased in primary human fibroblasts by 2.12× and 1.56× for 10-HSA and 9-HSA, respectively, in vitro with the10-HSA being significant (p < 0.05), whereas 12-HSA and SA had no statistical effect over the untreated control. 10-HSA and 12-HSA modified the intracellular fibroblast collagen proteome slightly with significant increases in collagen alpha-1 (VI) and alpha-3 (VI) proteins but only 10-HSA increased levels of collagen alpha-2 (V), alpha-1 (III), alpha-1 (I) and alpha-2 (I) (all p < 0.05) with the increases being significantly different between 10-HSA and 12-HSA for collagen alpha-1 (I), collagen-3 (VI) and collagen alpha-2 (I) (p < 0.01). Collagen type III in ex vivo skin was increased +47% (p < 0.05) by 0.05% (1.7 mM) retinol, +70% (p < 0.01) by 0.01% (0.33 mM) 10-HSA and the combination increased levels by +240% (p < 0.01 for either ingredient). CONCLUSION: Chiral (R)-10-HSA has been shown to be superior to 9, 12 and 17-HSA as a PPARα agonist. Moreover, 10-HSA stimulated collagen synthesis in monolayer fibroblast culture as assessed by proteomics and immunohistochemically. Furthermore, we also show the synergistic effects of 10-HSA with retinol on collagen III synthesis in skin explants. These results further highlight the efficacy of 10-HSA as a cosmetically acceptable PPARα agonist and anti-ageing ingredient.


INTRODUCTION: Nous rapportons les effets in vitro et ex vivo de l'acide chiral (R)-10-hydroxystéarique (10-HSA) par rapport à d'autres régioisomères d'acide mono-hydroxystéarique et à l'acide stéarique (SA) ainsi que ses avantages lorsqu'il est associé au rétinol. MÉTHODES: Après un traitement avec des acides hydroxystéariques, l'activité du récepteur alpha activé par les proliférateurs peroxysomaux (PPARα) a été déterminée dans un test du gène rapporteur de la luciférase, le collagène de type I a été évalué dans les fibroblastes dermiques humains primaires par immunohistochimie, la modification du protéome du collagène des fibroblastes intracellulaires a été étudiée par spectrométrie de masse. La protéomique et le collagène de type III ont été évalués par immunohistochimie sur la peau humaine ex vivo. RÉSULTATS: la 10-HSA était l'agoniste PPARα le plus efficace (induction 15,7X ; p<0,001), suivi de la 9-HSA (induction 10,1X) puis de la 12-HSA (induction 4,9X) avec la 17-HSA (induction 1,7X) étant similaire aux effets de l'acide stéarique (induction 1,8X). Les niveaux de collagène de type I ont été augmentés dans les fibroblastes humains primaires de 2,12X et 1,56X pour la 10-HSA et la 9-HSA respectivement in vitro, la 10-HSA étant significative (p<0,05) : alors que la 12-HSA et la SA n'ont eu aucun effet statistique sur le témoin non traité. La 10-HSA et la 12-HSA ont légèrement modifié le protéome du collagène des fibroblastes intracellulaires avec des augmentations significatives des protéines de collagène alpha-1 (VI) et alpha-3 (VI), mais seule la 10-HSA a augmenté les niveaux de collagène alpha-2 (V), alpha -1 (III), alpha-1 (I) et alpha-2 (I) (tous p<0,05) avec des augmentations significativement différentes entre 10-HSA et 12-HSA pour le collagène alpha-1 (I), le collagène- 3 (VI) et Collagène alpha-2 (I) (p<0,01). Le collagène de type III dans la peau ex vivo a augmenté de +47 % (p<0,05) de 0,05 % (1,7 mM) de rétinol, de +70 % (p<0,01) de 0,01 % (0,33 mM) de 10-HSA et la combinaison a augmenté les niveaux de +240 % (p<0,01 pour chaque ingrédient). CONCLUSION: La chiral (R)-10-HSA s'est avérée supérieure à 9, 12 et 17-HSA en tant qu'agoniste de PPARα. De plus, la 10-HSA a stimulé la synthèse de collagène dans la culture de fibroblastes monocouche telle qu'évaluée par protéomique et immunohistochimique. De plus, nous montrons également les effets synergiques de la 10-HSA avec le rétinol sur la synthèse du collagène III dans les explants de peau. Ces résultats soulignent en outre l'efficacité de la 10-HSA en tant qu'agoniste de PPARα et ingrédient anti-âge cosmétiquement acceptable.


Subject(s)
Collagen Type III/drug effects , Collagen Type I/drug effects , PPAR alpha/pharmacology , Retinoids/pharmacology , Skin Aging/drug effects , Stearic Acids/pharmacology , Drug Synergism , Female , Fibroblasts/drug effects , HEK293 Cells , Humans
2.
Article in English | MEDLINE | ID: mdl-34246925

ABSTRACT

The additive effects of prostaglandin (PG)-EP2 agonists on a PG-FP agonist toward adipogenesis in two- or three-dimension (2D or 3D) cultures of 3T3-L1 cells was examined by lipid staining, the mRNA expression of adipogenesis related genes, and extracellular matrixes (ECMs) including collagen molecules (Col) -1, -4 and -6, and fibronectin (Fn), and the sizes and physical properties of 3D sphenoids, as measured by a micro-squeezer. The results indicate that adipogenesis induced 1) an enlargement in the sizes of 3D sphenoids, 2) a substantial enhancement in lipid staining, the expression of the PParγ, Ap2 and Leptin genes, and 3) a significant decrease in the stiffness of the 3D sphenoids. These effects were inhibited by bimatoprost acid (BIM-A), but 4) adipogenesis induced significant down-regulation of Col1 and Fn, and the significant up-regulation of the Col4 and Col6 genes were unchanged by BIM-A. On the addition of an EP2 agonist, such as omidenepag (OMD) or butaprost (Buta), to BIM-A, 1) the sizes of the 3D sphenoids were further decreased, 2) lipid staining was decreased (2D; OMD, 3D; Buta) 3) the stiffness of the 3D sphenoids was increased by Buta, 4) the expression of PParγ was up-regulated (2D; Buta) or unchanged (3D), the expression of Ap2 was down-regulated (2D; OMD) or up-regulated (3D; Buta), and the expression of Leptin was increased (2D), 5) the expression of all four (OMD) or all except Col4 (buta) in 2D, and Col1and Col4 (OMD) in 3D were up-regulated. These collective findings indicate that the addition of an EP2 agonist, OMD or Buta significantly modulated the BIM-A induced suppression of adipogenesis as well as physical properties of 2D and 3D cultured 3T3-L1 cells in different manners.


Subject(s)
Adipogenesis/drug effects , Alprostadil/analogs & derivatives , Bimatoprost/pharmacology , Fatty Acid-Binding Proteins/drug effects , Glycine/analogs & derivatives , Leptin/genetics , PPAR gamma/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin/agonists , 3T3-L1 Cells , Adipogenesis/genetics , Alprostadil/pharmacology , Animals , Cell Culture Techniques, Three Dimensional , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type IV/drug effects , Collagen Type IV/genetics , Collagen Type IV/metabolism , Collagen Type VI/drug effects , Collagen Type VI/genetics , Collagen Type VI/metabolism , Drug Synergism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fibronectins/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Glycine/pharmacology , Leptin/metabolism , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
3.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069423

ABSTRACT

Endometrosis is a reproductive pathology that is responsible for mare infertility. Our recent studies have focused on the involvement of neutrophil extracellular traps enzymes, such as elastase (ELA), in the development of equine endometrosis. Noscapine (NOSC) is an alkaloid derived from poppy opium with anticough, antistroke, anticancer, and antifibrotic properties. The present work investigates the putative inhibitory in vitro effect of NOSC on collagen type I alpha 2 chain (COL1A2) mRNA and COL1 protein relative abundance induced by ELA in endometrial explants of mares in the follicular or mid-luteal phases at 24 or 48 h of treatment. The COL1A2 mRNA was evaluated by qPCR and COL1 protein relative abundance by Western blot. In equine endometrial explants, ELA increased COL 1 expression, while NOSC inhibited it at both estrous cycle phases and treatment times. These findings contribute to the future development of new endometrosis treatment approaches. Noscapine could be a drug capable of preventing collagen synthesis in mare's endometrium and facilitate the therapeutic approach.


Subject(s)
Collagen Type I/metabolism , Endometriosis/metabolism , Noscapine/pharmacology , Animals , Collagen/metabolism , Collagen Type I/drug effects , Collagen Type I/genetics , Endometriosis/drug therapy , Endometriosis/veterinary , Endometrium/drug effects , Endometrium/metabolism , Estrous Cycle , Extracellular Traps/metabolism , Female , Fibrosis , Horse Diseases/pathology , Horses , Noscapine/metabolism , Pancreatic Elastase/metabolism , Protease Inhibitors/pharmacology
4.
Biomolecules ; 11(5)2021 04 21.
Article in English | MEDLINE | ID: mdl-33919331

ABSTRACT

Stress is a major contributing factor of skin aging, which is clinically characterized by wrinkles, loss of elasticity, and dryness. In particular, glucocorticoids are generally considered key hormones for promoting stress-induced skin aging through binding to glucocorticoid receptors (GRs). In this work, we aimed to investigate whether ß-ionone (a compound occurring in various foods such as carrots and almonds) attenuates dexamethasone-induced suppression of collagen and hyaluronic acid synthesis in human dermal fibroblasts, and to explore the mechanisms involved. We found that ß-ionone promoted collagen production dose-dependently and increased mRNA expression levels, including collagen type I α 1 chain (COL1A1) and COL1A2 in dexamethasone-treated human dermal fibroblasts. It also raised hyaluronic acid synthase mRNA expression and hyaluronic acid levels. Notably, ß-ionone inhibited cortisol binding to GR, subsequent dexamethasone-induced GR signaling, and the expression of several GR target genes. Our results reveal the strong potential of ß-ionone for preventing stress-induced skin aging and suggest that its effects are related to the inhibition of GR signaling in human dermal fibroblasts.


Subject(s)
Norisoprenoids/metabolism , Norisoprenoids/pharmacology , Skin/metabolism , Aging/drug effects , Aging/metabolism , Cell Line , Cells, Cultured , Collagen/drug effects , Collagen/metabolism , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Dexamethasone/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Hyaluronic Acid/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin Aging/drug effects , Transforming Growth Factor beta/metabolism , Ultraviolet Rays
5.
Eur J Endocrinol ; 185(1): 33-45, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33886495

ABSTRACT

OBJECTIVE: The insulinotropic effect of exogenous, intravenously infused glucose-dependent insulinotropic polypeptide (GIP) is impaired in patients with type 2 diabetes. We evaluated the effects of endogenous GIP in relation to glucose and bone metabolism in patients with type 2 diabetes using a selective GIP receptor antagonist and hypothesized that the effects of endogenous GIP were preserved. DESIGN: A randomized, double-blinded, placebo-controlled, crossover study. METHODS: Ten patients with overweight/obesity and type 2 diabetes (mean±s.d.; HbA1c 52 ± 11 mmol/mol; BMI 32.5 ± 4.8 kg/m2) were included. We infused a selective GIP receptor antagonist, GIP(3-30)NH2 (1200 pmol/kg/min), or placebo (saline) during two separate, 230-min, standardized, liquid mixed meal tests followed by a meal ad libitum. Subcutaneous adipose tissue biopsies were analyzed. RESULTS: Compared with placebo, GIP(3-30)NH2 reduced postprandial insulin secretion (Δbaseline-subtracted area under the curve (bsAUC)C-peptide% ± s.e.m.; -14 ± 6%, P = 0.021) and peak glucagon (Δ% ± s.e.m.; -11 ± 6%, P = 0.046) but had no effect on plasma glucose (P = 0.692). Suppression of bone resorption (assessed by circulating carboxy-terminal collagen crosslinks (CTX)) was impaired during GIP(3-30)NH2 infusion compared with placebo (ΔbsAUCCTX; ±s.e.m.; -4.9 ± 2 ng/mL × min, P = 0.005) corresponding to a ~50% reduction. Compared with placebo, GIP(3-30)NH2 did not affect plasma lipids, meal consumption ad libitum or adipose tissue triglyceride content. CONCLUSIONS: Using a selective GIP receptor antagonist during a meal, we show that endogenous GIP increases postprandial insulin secretion with little effect on postprandial glycaemia but is important for postprandial bone homeostasis in patients with type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Bone Resorption/metabolism , Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/metabolism , Insulin Secretion/physiology , Obesity/metabolism , Triglycerides/metabolism , Adult , Aged , Blood Glucose/drug effects , Collagen Type I/drug effects , Collagen Type I/metabolism , Cross-Over Studies , Double-Blind Method , Feeding Behavior/drug effects , Gastric Inhibitory Polypeptide/pharmacology , Humans , Insulin Secretion/drug effects , Male , Middle Aged , Peptide Fragments/pharmacology , Peptides/drug effects , Peptides/metabolism , Postprandial Period , Random Allocation , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism
6.
Eur J Clin Invest ; 51(7): e13534, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33656763

ABSTRACT

BACKGROUND: Statins are 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors blocking cholesterol biosynthesis in hepatic cells, thereby causing an increase in low-density lipoprotein (LDL) receptors resulting in enhanced uptake and clearance of atherogenic LDL-cholesterol (LDL-C) from the blood. Accordingly, statins decrease the risk of developing atherosclerosis and its acute complications, such as acute myocardial infarction and ischaemic stroke. Besides the LDL-C-lowering impact, statins also have other so-called pleiotropic effects. Among them, the ability to modulate differentiation and function of bone cells and exert direct effects on osteosynthesis factors. Specifically, earlier studies have shown that statins cause in vitro and in vivo osteogenic differentiation. DESIGN: The most relevant papers on the bone-related 'pleiotropic' effects of statins were selected following literature search in databases and were reveiwed. RESULTS: Statins increase the expression of many mediators involved in bone metabolism including bone morphogenetic protein-2 (BMP-2), glucocorticoids, transforming growth factor-beta (TGF-ß), alkaline phosphatase (ALP), type I collagen and collagenase-1. As a result, they enhance bone formation and improve bone mineral density by modulating osteoblast and osteoclast differentiation. CONCLUSION: This review summarizes the literature exploring bone-related 'pleiotropic' effects of statins and suggests an anabolic role in the bone tissue for this drug class. Accordingly, current knowledge encourages further clinical trials to assess the therapeutic potential of statins in the treatment of bone disorders, such as arthritis and osteoporosis.


Subject(s)
Bone Density/drug effects , Cell Differentiation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 2/metabolism , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagenases/drug effects , Collagenases/metabolism , Glucocorticoids/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
7.
Int J Mol Sci ; 22(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572500

ABSTRACT

Collagen type I is a key structural component of dermis tissue and is produced by fibroblasts and the extracellular matrix. The skin aging process, which is caused by intrinsic or extrinsic factors, such as natural aging or free radical exposure, greatly reduces collagen expression, thereby leading to obstructed skin elasticity. We investigated the effective fermentation of Cetearyl isononanoate (CIN), a polyethylene glycol (PEG) analog, as a carbon source with the skin probiotic bacterium Staphylococcus epidermidis (S.epidermidis) or butyrate, as their fermentation metabolites could noticeably restore collagen expression through phosphorylated extracellular signal regulated kinase (p-ERK) activation in mouse fibroblast cells and skin. Both the in vitro and in vivo knockdown of short-chain fatty acid (SCFA) or free fatty acid receptor 2 (FFaR2) considerably blocked the probiotic effect of S. epidermidis on p-ERK-induced collagen type I induction. These results demonstrate that butyric acid (BA) in the metabolites of fermenting skin probiotic bacteria mediates FFaR2 to induce the synthesis of collagen through p-ERK activation. We hereby imply that metabolites from the probiotic S. epidermidis fermentation of CIN as a potential carbon source could restore impaired collagen in the dermal extracellular matrix (ECM), providing integrity and elasticity to skin.


Subject(s)
Butyric Acid/metabolism , Collagen Type I/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Probiotics , Receptors, G-Protein-Coupled/metabolism , Skin Aging/drug effects , Staphylococcus epidermidis/physiology , Animals , Collagen Type I/drug effects , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Fatty Acids, Volatile/metabolism , Female , Fermentation , Fibroblasts , MAP Kinase Signaling System/drug effects , Mice , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Skin/metabolism , Skin/microbiology
8.
Gynecol Endocrinol ; 37(4): 342-348, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33480297

ABSTRACT

OBJECTIVE: To investigate the therapeutic effects of PERK activator CCT020312 (CCT) on inflammation-mediated osteoporosis (IMO) in ovariectomized rats. METHODS: Rats were divided into Sham, IMO, IMO + 1 mg/kg CCT and IMO + 2 mg/kg CCT groups. IMO models were constructed by bilateral ovariectomy (OVX) on 1st day followed by injection with magnesium silicate (Talc) on the 59th day. Sham rats did not undergo OVX surgery and were injected with saline instead of Talc. From 60th to 79th day, rats were treated with DMSO (vehicle control) in the Sham and IMO groups, and 1 or 2 mg/kg CCT020312 in treatment groups. Osteopontin (OPN), osteocalcin (OCN), tartrate-resistant acid phosphatase (TRAP), C-terminal telopeptide of type I collagen (CTX-I), and pro-inflammatory factors were measured on the 80th day. ProdigyDEXA was used to evaluate bone mineral density and content (BMD/BMC). Bone volume/total volume (BV/TV), connectivity density (Conn.D), trabecular number (Tb.N), and trabecular separation (Tb.Sp) was assessed using 3D micro-CT scanner. RESULTS: CCT up-regulated Conn.D, BV/TV, and Tb.N, but down-regulated Tb.Sp in IMO rats. Besides, the declined femoral BMD and BMC in IMO rats were elevated after CCT treatment. Besides, IMO rats represented declined OPN and OCN, as well as increased TRAP, CTX-I, and pro-inflammatory factors, whereas those in the treatment groups were ameliorated regarding these indexes, with 2 mg/kg CCT showing better effect. CONCLUSION: PERK activator CCT020312 can be served as a new therapeutic option for the protection against bone loss in the OVX rat model associated with inflammation probably by manipulating inflammatory factors.


Subject(s)
Bone Density/drug effects , Bone and Bones/drug effects , Enzyme Activators/pharmacology , Ovariectomy , eIF-2 Kinase , Absorptiometry, Photon , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Cancellous Bone/diagnostic imaging , Cancellous Bone/drug effects , Collagen Type I/drug effects , Collagen Type I/metabolism , Femur/diagnostic imaging , Femur/drug effects , Femur/metabolism , Humans , Imaging, Three-Dimensional , Inflammation/metabolism , Organ Size , Osteocalcin/drug effects , Osteocalcin/metabolism , Osteopontin/drug effects , Osteopontin/metabolism , Osteoporosis, Postmenopausal/diagnostic imaging , Osteoporosis, Postmenopausal/metabolism , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Rats , Tartrate-Resistant Acid Phosphatase/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism , X-Ray Microtomography
9.
Endocr J ; 68(3): 307-315, 2021 Mar 28.
Article in English | MEDLINE | ID: mdl-33115984

ABSTRACT

Testosterone deficiency is associated with poor prognosis among patients with chronic heart failure (HF). Physiological testosterone improves the exercise capacity of patients with HF. In this study, we evaluated whether treatment with physiological testosterone contributes to anti-fibrogenesis by modifying calcium homeostasis in cardiac fibroblasts and we studied the underlying mechanisms. Nitric oxide (NO) analyses, calcium (Ca2+) fluorescence, and Western blotting were performed in primary isolated rat cardiac fibroblasts with or without (control cells) testosterone (10, 100, 1,000 nmol/L) treatment for 48 hours. Physiological testosterone (10 nmol/L) increased NO production and phosphorylation at the inhibitory site of the inositol trisphosphate (IP3) receptor, thereby reducing Ca2+ entry, phosphorylated Ca2+/calmodulin-dependent protein kinase II (CaMKII) expression, type I and type III pro-collagen production. Non-physiological testosterone-treated fibroblasts exhibited similar NO and collagen production capabilities as compared to control (testosterone deficient) fibroblasts. These effects were blocked by co-treatment with NO inhibitor (L-NG-nitro arginine methyl ester [L-NAME], 100 µmol/L). In the presence of the IP3 receptor inhibitor (2-aminoethyl diphenylborinate [2-APB], 50 µmol/L), testosterone-deficient and physiological testosterone-treated fibroblasts exhibited similar phosphorylated CaMKII expression. When treated with 2-APB or CaMKII inhibitor (KN93, 10 µmol/L), testosterone-deficient and physiological testosterone-treated fibroblasts exhibited similar type I, and type III collagen production. In conclusion, physiological testosterone activates NO production, and attenuates the IP3 receptor/Ca2+ entry/CaMKII signaling pathway, thereby inhibiting the collagen production capability of cardiac fibroblasts.


Subject(s)
Androgens/pharmacology , Calcium/metabolism , Fibroblasts/drug effects , Nitric Oxide/metabolism , Testosterone/pharmacology , Androgens/physiology , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinase Type 2/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type III/drug effects , Collagen Type III/metabolism , Fibroblasts/metabolism , Fibrosis , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Myocardium/cytology , Rats , Testosterone/physiology
10.
Aging (Albany NY) ; 13(4): 4976-4985, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33196456

ABSTRACT

Osteogenic differentiation is critical to bone homeostasis, and its imbalance plays a key role in the progression of osteoporosis. Osteoblast cells are responsible for synthesizing new bone tissue, and understanding how to control osteoblastic differentiation is vital to the treatment of osteoporosis. Herein, we show that GPR173 signaling is involved in the regulation of osteoblastic differentiation in MC3T3-E1 cells. Our data reveals that GPR173 is abundantly expressed in MC3T3-E1 cells, and its expression is inducible upon the introduction of osteogenic media. The activation of GPR173 by its selective agonist phoenixin 20 induces the expression of several osteoblast signature genes including collagen type 1 alpha 1 (Col-I), osteocalcin (OCN), alkaline phosphatase (ALP) as well as increased matrix mineralization and ALP activity, suggesting that the activation of GPR173 promotes osteoblastic differentiation. Moreover, we show that the effect of phoenixin 20 is mediated by its induction on the key regulator runt-Related Transcription Factor 2 (Runx2). Mechanistically, we display that the action of phoenixin 20 requires the activation of MAPK kinase p38, and deactivation of p38 by its inhibitor SB203580 weakens the phoenixin 20-mediated induction of RUNX-2, ALP, and matrix mineralization. Silencing of GPR173 attenuates phoenixin 20-mediated osteoblastic differentiation, indicating its dependence on the receptor. Collectively, our study reveals a new role of GPR173 and its agonist phoenixin 20 in osteoblastic differentiation.


Subject(s)
Calcification, Physiologic/genetics , Cell Differentiation/physiology , Osteoblasts/physiology , Receptors, G-Protein-Coupled/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic/drug effects , Calcification, Physiologic/physiology , Cell Differentiation/drug effects , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Enzyme Inhibitors/pharmacology , Gene Expression , Gene Knockdown Techniques , Imidazoles/pharmacology , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteocalcin/genetics , Peptide Hormones/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Transcriptome , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Medicine (Baltimore) ; 99(40): e22542, 2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33019463

ABSTRACT

BACKGROUND: The goal of this study was to review relevant randomized controlled trials or case-control studies to determine the clinical efficacy of minodronate in the treatment of osteoporosis. METHOD: The relevant studies were identified on PubMed, Cochrane, and Embase databases using appropriate keywords. Pertinent sources in the literature were also reviewed, and all articles published through October 2019 were considered for inclusion. For each study, we assessed odds ratios, mean difference, and 95% confidence interval (95% CI) to evaluate and synthesize outcomes. RESULT: Thirteen studies comprising 3740 patients were included in this study. Compared with other drugs, minodronate significantly decreased N-telopeptide of type I collagen/creatinine (weighted mean difference [WMD]: -13.669, 95% confidence interval [CI]: -23.108 to -4.229), bone alkaline phosphatase (BAP) (WMD: -1.26, 95% CI: -2.04 to -0.47) and tartrate-resistant acid phosphatase 5b (WMD: -154.11, 95% CI: -277.85 to -30.37). Minodronate combined with other drugs would significantly decrease BAP (WMD: -3.10, 95% CI: -5.20 to -1.00) than minodronate. Minodronate-naïve would significantly decrease BAP (WMD: -3.00, 95% CI: -5.47 to 0.53) and tartrate-resistant acid phosphatase 5b (WMD: -128.20, 95% CI: -198.11 to -58.29) than minodronate-switch. The incidence of vertebral fracture was significantly decreased in the minodronate group than the other drugs (relative risk: 0.520, 95% CI: 0.363-0.744). CONCLUSION: Minodronate has better clinical efficacy in the treatment of osteoporosis than other drugs (alendronate, risedronate, raloxifene, or eldecalcitol).


Subject(s)
Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Osteoporosis/drug therapy , Aged , Aged, 80 and over , Alendronate/therapeutic use , Alkaline Phosphatase/drug effects , Bone Density Conservation Agents/therapeutic use , Case-Control Studies , Collagen Type I/drug effects , Creatinine , Drug Therapy, Combination/statistics & numerical data , Female , Humans , Male , Middle Aged , Raloxifene Hydrochloride/therapeutic use , Randomized Controlled Trials as Topic , Risedronic Acid/therapeutic use , Spinal Fractures/epidemiology , Tartrate-Resistant Acid Phosphatase/drug effects , Treatment Outcome , Vitamin D/analogs & derivatives , Vitamin D/therapeutic use
12.
PLoS One ; 15(7): e0236992, 2020.
Article in English | MEDLINE | ID: mdl-32735603

ABSTRACT

We wanted to investigate whether Isoniazid (INH) can directly stimulate activation of hepatic stellate cells (HSCs) and enhance production of collagen. Treatment of human hepatic stellate cell line LX2 with or without 5µM INH for 24 to 72 hours was performed to look into content of cytochrome P450 2E1 (CYP2E1), activity of NADPH oxidase (NOX) and intracellular oxidative stress. Protein level as well as mRNA expression of alpha smooth muscle actin (α-SMA) and collagen1A1 (COL1A1) were assessed by western blot and real time PCR. In some experiments pyrazole (PY) was pre-treated to LX2 cells to induce CYP2E1 prior to INH treatment. CYP2E1 level as well as NOX activity was gradually increased with INH treatment in LX2 cells till 72 hours. Following 72 hours of INH exposure, intracellular glutathione (GSH) level was found to be reduced compared to control (p<0.01) and showed expression of α-SMA, indicating activation of HSC. We could not found any change in collagen expression in this experimental study. Pyrazole (PY) pre-treatment to LX2 cells caused significant increase in cellular CYP2E1 content associated with increase of NOX, intracellular reactive oxygen species (ROS), and expression of α-SMA and collagen1 after INH exposure. CYP2E1 is present in insignificant amount in HSCs and INH treatment could not induce collagen expression, although altered cellular oxidant levels was observed. But in LX2 cells when CYP2E1 was over-expressed by PY, INH administration provokes oxidative stress mediated stellate cells activation along with collagen type I expression.


Subject(s)
Collagen Type I , Cytochrome P-450 CYP2E1 , Isoniazid/pharmacology , Cell Line , Collagen Type I/drug effects , Collagen Type I/metabolism , Cytochrome P-450 CYP2E1/drug effects , Cytochrome P-450 CYP2E1/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , NADPH Oxidases/drug effects , Oxidative Stress/drug effects
13.
Int J Biol Macromol ; 164: 748-758, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32693139

ABSTRACT

Artificial sweeteners (AS) are widely used as sugar substitutes because natural sweetener (sugar) leads to a number of health issues, including diabetes, obesity, and tooth decay. Since natural sugar (sucrose), diabetes and skin are highly interlinked, and also sucrose is known to inhibit the fibrillation of collagen, the major protein of the skin, a study on the impact of AS on collagen is important and essential. Herein, we have studied the influence of commonly used AS such as Sucralose (SUC), Aspartame (APM), and Saccharin (SAC) on the structure, stability, and fibrillation of collagen using various spectroscopic methods. The circular dichroism and turbidity results suggest that the AS does not disrupt the triple helix structure and also the fibrillar property of collagen, respectively. The fibrillar morphology was sustained, although there was a trivial difference in the entanglement of fibrils in the presence of SAC, compared to native collagen fibrils. The thermal stability of collagen is maintained in the presence of AS. Fluorescence and STD-NMR results indicate that the interaction between AS and collagen was weak, which supports the intact structure, stability, and fibrillation property of collagen. The current study thus suggests that the chosen AS does not influence collagen properties.


Subject(s)
Collagen Type I/ultrastructure , Fibril-Associated Collagens/ultrastructure , Sucrose/adverse effects , Sweetening Agents/pharmacology , Aspartame/adverse effects , Aspartame/pharmacology , Collagen Type I/chemistry , Collagen Type I/drug effects , Diabetes Mellitus/epidemiology , Fibril-Associated Collagens/chemistry , Fibril-Associated Collagens/drug effects , Humans , Obesity/epidemiology , Saccharin/adverse effects , Saccharin/pharmacology , Sucrose/analogs & derivatives , Sucrose/pharmacology , Sweetening Agents/adverse effects
14.
Acta Cir Bras ; 35(3): e202000303, 2020.
Article in English | MEDLINE | ID: mdl-32490900

ABSTRACT

PURPOSE: To evaluate the in vivo response of photobiomodulation therapy associated with norbixin-based poly(hydroxybutyrate) membrane (PHB) in tenotomized calcaneal tendon. METHODS: Thirty rats were randomly allocated to six groups (n=5 each): LED groups (L1, L2 and L3) and membrane + LED groups (ML1, ML2 and ML3). The right calcaneal tendons of all animals were sectioned transversely and were irradiated with LED daily, one hour after surgery every 24 hours, until the day of euthanasia. At the end of the experiments the tendons were removed for histological analysis. RESULTS: The histological analysis showed a significant reduction in inflammatory cells in the ML1, ML2 and ML3 groups (p=0.0056, p=0.0018 and p<0.0001, respectively) compared to those in the LED group. There was greater proliferation of fibroblasts in the ML1 (p<0.0001) and L3 (p<0.0001) groups. A higher concentration of type I collagen was also observed in the ML1 group (p=0.0043) replacing type III collagen. CONCLUSION: Photobiomodulation in association with norbixin-based PHB membrane led to control of the inflammatory process. However, it did not favor fibroblast proliferation and did not optimize type I collagen formation in the expected stage of the repair process.


Subject(s)
Achilles Tendon/radiation effects , Carotenoids/pharmacology , Hydroxybutyrates/pharmacology , Low-Level Light Therapy/methods , Tendinopathy/radiotherapy , Tenotomy/methods , Achilles Tendon/drug effects , Achilles Tendon/surgery , Animals , Collagen/pharmacology , Collagen Type I/analysis , Collagen Type I/drug effects , Collagen Type III/analysis , Collagen Type III/drug effects , Drug Evaluation, Preclinical , Fibroblasts/chemistry , Fibroblasts/drug effects , Male , Prohibitins , Random Allocation , Rats , Rats, Wistar , Wound Healing/drug effects , Wound Healing/radiation effects
15.
Pediatr Rheumatol Online J ; 18(1): 46, 2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32517762

ABSTRACT

BACKGROUND: The effects of C-type natriuretic peptide (CNP) and fibroblast growth factor (FGF)-23 appear to oppose each other during the process of bone formation, whereas few studies exist on the interaction between CNP and FGF-23. The main objective of the present study is to probe whether CNP is directly responsible for the regulation of osteoblast or via antagonizing FGF-23. METHODS: Osteoblasts were cultured in the absence or presence of CNP (0, 10, and 100 pmol/L) for 24 h, 48 h and 72 h, respectively. RESULTS: The findings of the present study indicated that: (1) CNP significantly stimulated osteoblastic proliferation and collagen (Col)-X expression; (2) both osteoblastic (osteocalcin, procollagen type I carboxy-terminal propeptide, total alkaline phosphatase and bone-specific alkaline phosphatase) and osteolytic (tartrate-resistant acid phosphatase and cross-linked carboxyterminal telopeptide of type I collagen) bone turnover biomarkers were up-regulated by CNP in osteoblasts; (3) FGF-23 mRNA and protein were significantly down-regulated at 24 h by CNP in osteoblasts, but the expression of FGF receptor-1/Klotho had no significant change. CONCLUSIONS: CNP stimulates osteoblastic proliferation and Col-X expression via the down-regulation of FGF-23 possibly in vitro. However, the specific mechanisms of the interaction between CNP and FGF-23 in osteoblasts are still unclear according to our findings. A further study on osteoblasts cultured with CNP and FGF-23 inhibitor will be undertaken in our laboratory.


Subject(s)
Cell Proliferation/genetics , Fibroblast Growth Factors/genetics , Natriuretic Peptide, C-Type/metabolism , Osteoblasts/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Bone Remodeling/drug effects , Bone Remodeling/genetics , Cell Proliferation/drug effects , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type X/drug effects , Collagen Type X/genetics , Collagen Type X/metabolism , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/drug effects , Fibroblast Growth Factors/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation , Glucuronidase/drug effects , Glucuronidase/genetics , Glucuronidase/metabolism , In Vitro Techniques , Klotho Proteins , Natriuretic Peptide, C-Type/pharmacology , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteocalcin/metabolism , Osteogenesis/genetics , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Primary Cell Culture , Procollagen/drug effects , Procollagen/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/drug effects , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Tartrate-Resistant Acid Phosphatase/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism
16.
Int J Pharm ; 585: 119470, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32464234

ABSTRACT

The aim of this study was to prepare and evaluate simvastatin (SIM) loaded elastic provesicular systems for effective topical wound management. SIM provesicles were prepared using the non-ionic surfactant Span 40, cholesterol and three edge activators i.e. Span 80, Tween 80 and sodium cholate. The vesicles revealed high SIM encapsulation efficiency ranging from 87.25 to 98.15%, whereas vesicle sizes ranged from 462.3 to 801.5 nm. Vesicle sizes decreased with increasing the concentration of the edge activator. High negative zeta potential values were observed, revealing good stability of the vesicular formulations. The release of SIM from hydrated provesicular carriers was biphasic in nature. The selected SIM provesicular elastic carrier exerted approximately two-fold increase in the amount of SIM permeated through rat skin, compared to the free drug. Evaluation of wound healing activity of the selected provesicular formulation revealed significant reduction in wound size in rats, fourteen days post-wounding. These results were further confirmed by a significant increase in expression of vascular endothelial growth factor and collagen type I compared to the free drug. These results indicate that provesicular carriers could be a promising drug delivery system for encapsulating SIM and enhancing its wound healing efficacy.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Carriers/pharmacology , Simvastatin/pharmacology , Wound Healing/drug effects , Animals , Cholesterol/chemistry , Collagen Type I/drug effects , Drug Carriers/chemistry , Drug Liberation , Drug Stability , Hexoses/chemistry , Male , Particle Size , Polysorbates/chemistry , Rats , Rats, Wistar , Simvastatin/administration & dosage , Sodium Cholate/chemistry , Vascular Endothelial Growth Factor A/drug effects
17.
Am J Obstet Gynecol ; 223(5): 733.e1-733.e14, 2020 11.
Article in English | MEDLINE | ID: mdl-32417359

ABSTRACT

BACKGROUND: Uterine leiomyomas, the most common tumors of the female reproductive system, are characterized by excessive deposition of disordered stiff extracellular matrix and fundamental alteration in the mechanical signaling pathways. Specifically, these alterations affect the normal dynamic state of responsiveness to mechanical cues in the extracellular environment. These mechanical cues are converted through integrins, cell membrane receptors, to biochemical signals including cytoskeletal signaling pathways to maintain mechanical homeostasis. Leiomyoma cells overexpress ß1 integrin and other downstream mechanical signaling proteins. We previously reported that simvastatin, an antihyperlipidemic drug, has antileiomyoma effects through cellular, animal model, and epidemiologic studies. OBJECTIVE: This study aimed to examine the hypothesis that simvastatin might influence altered mechanotransduction in leiomyoma cells. STUDY DESIGN: This is a laboratory-based experimental study. Primary leiomyoma cells were isolated from 5 patients who underwent hysterectomy at the Department of Gynecology and Obstetrics of the Johns Hopkins University Hospital. Primary and immortalized human uterine leiomyoma cells were treated with simvastatin at increasing concentrations (0.001, 0.01, 0.1, and 1 µM, or control) for 48 hours. Protein and mRNA levels of ß1 integrin and extracellular matrix components involved in mechanical signaling were quantified by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. In addition, we examined the effect of simvastatin on the activity of Ras homolog family member A using pull-down assay and gel contraction. RESULTS: We found that simvastatin significantly reduced the protein expression of ß1 integrin by 44% and type I collagen by 60% compared with untreated leiomyoma cells. Simvastatin-treated cells reduced phosphorylation of focal adhesion kinase down to 26%-60% of control, whereas it increased total focal adhesion kinase protein expression. Using a Ras homolog family member A pull-down activation assay, we observed reduced levels of active Ras homolog family member A in simvastatin-treated cells by 45%-85% compared with control. Consistent with impaired Ras homolog family member A activation, simvastatin treatment reduced tumor gel contraction where gel area was 122%-153% larger than control. Furthermore, simvastatin treatment led to reduced levels of mechanical signaling proteins involved in ß1 integrin downstream signaling, such as A-kinase anchor protein 13, Rho-associated protein kinase 1, myosin light-chain kinase, and cyclin D1. CONCLUSION: The results of this study suggest a possible therapeutic role of simvastatin in restoring the altered state of mechanotransduction signaling in leiomyoma. Collectively, these findings are aligned with previous epidemiologic studies and other reports and support the need for clinical trials.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Leiomyoma/genetics , Mechanotransduction, Cellular/drug effects , Simvastatin/pharmacology , Uterine Neoplasms/genetics , A Kinase Anchor Proteins/drug effects , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Cyclin D1/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/drug effects , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin beta1/drug effects , Integrin beta1/genetics , Integrin beta1/metabolism , Leiomyoma/metabolism , Mechanotransduction, Cellular/genetics , Minor Histocompatibility Antigens/drug effects , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Myosin-Light-Chain Kinase/drug effects , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , Primary Cell Culture , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Uterine Neoplasms/metabolism , rho-Associated Kinases/drug effects , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
18.
Wound Repair Regen ; 28(4): 460-469, 2020 07.
Article in English | MEDLINE | ID: mdl-32428986

ABSTRACT

Hypertrophic scar is an important clinical problem with limited therapeutic options. Aside from their roles as 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors, statins have also been demonstrated to decrease scarring by reducing connective tissue growth factor (CTGF) expression. However, poor penetrative ability limits their utility as topical treatments for hypertrophic scar. Here, we aim to develop novel statin formulations using liposomes to enhance dermal penetrative ability and to evaluate their efficacy against formation of hypertrophic scar utilizing our validated rabbit ear hypertrophic scar model. Liposomal simvastatin or pravastatin were compounded using a novel, flexible liposomal formulation and applied topically to rabbit ear hypertrophic scars daily from postoperation day (POD) 14 until POD 25. Scar color, including erythema and melanin, was measured using reflectance spectrophotometry on POD 28, and scar tissue was harvested for evaluation of scar elevation index as well as gene and protein expression. Human foreskin fibroblasts were also treated with statin formulations and CCN2 expression was determined by quantitative PCR. Both simvastatin and pravastatin were efficiently encapsulated in liposomes, forming nanometer-scale particles possessing highly negative charges. Topical treatment with liposomal simvastatin and pravastatin at 6.5% concentration significantly reduced scar elevation index and decreased type I/III collagen content and myofibroblast persistence in the wound. The erythema/vascularity of scars was reduced by liposomal statin treatment, with concomitant decrease of CD31 expression as measured histologically. Expression levels of transcripts encoding CTGF, collagen I, and collagen III collagen in scar tissue were also decreased by liposomal pravastatin treatment, as were myofibroblast persistence and the type I/III collagen ratio as assessed by immunofluorescence and picrosirus red staining, respectively. Treatment of human foreskin fibroblasts with simvastatin or with liposome-encapsulated pravastatin resulted in decreased expression of transcript encoding CTGF. Overall, our novel statin formulations encapsulated in liposomes were successfully delivered through topical application, significantly reducing hypertrophic scarring in a rabbit ear model.


Subject(s)
Cicatrix, Hypertrophic/metabolism , Fibroblasts/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Skin/metabolism , Animals , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/prevention & control , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type III/drug effects , Collagen Type III/genetics , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/genetics , Ear, External/injuries , Ear, External/metabolism , Ear, External/pathology , Erythema , Fibroblasts/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , In Vitro Techniques , Liposomes , Melanins , Platelet Endothelial Cell Adhesion Molecule-1/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pravastatin/administration & dosage , Pravastatin/pharmacology , Rabbits , Simvastatin/administration & dosage , Simvastatin/pharmacology , Skin/injuries , Skin/pathology , Spectrophotometry
19.
Aging (Albany NY) ; 12(3): 2226-2245, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32023550

ABSTRACT

Osteoporosis is a metabolic disease characterized by reduced osteoblast differentiation and proliferation. Oxidative stress plays a role in the pathogenesis of osteoporosis. Aucubin (AU), an iridoid glycoside, was previously shown to promote osteoblast differentiation. We investigated the effects of AU on MG63 human osteoblast-like cells treated with dexamethasone (Dex) or hydrogen peroxide (H2O2) to induce oxidative damage. AU protected MG63 cells against apoptosis, and promoted increased expression of cytokines associated with osteoblast differentiation, including collagen I, osteocalcin (OCN), osteopontin (OPN), and osterix. In Dex- and H2O2-treated MG63 cells, AU also enhanced the expression of anti-oxidative stress-associated factors in the nuclear respiratory factor 2 signaling pathway, including superoxide dismutases 1 and 2, heme oxygenases 1 and 2, and catalase. In vivo, using a Dex-induced mouse model of osteoporosis, AU promoted increased cortical bone thickness, increased bone density, and tighter trabecular bone. Additionally, it stimulated an increase in the expression of collagen I, OCN, OPN, osterix, and phosphorylated Akt and Smads in bone tissue. Finally, AU stimulated the expression of cytokines associated with osteoblast differentiation in bone tissue and serum. Our data indicate AU may have therapeutic efficacy in osteoporosis.


Subject(s)
Bone and Bones/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Iridoid Glucosides/pharmacology , Osteoblasts/drug effects , Osteoporosis/metabolism , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line , Collagen Type I/drug effects , Collagen Type I/metabolism , Cytokines/drug effects , Cytokines/metabolism , Dexamethasone/adverse effects , GA-Binding Protein Transcription Factor/drug effects , GA-Binding Protein Transcription Factor/metabolism , Glucocorticoids/adverse effects , Humans , Hydrogen Peroxide/toxicity , Mice , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/drug effects , Osteocalcin/metabolism , Osteopontin/drug effects , Osteopontin/metabolism , Osteoporosis/chemically induced , Osteoporosis/pathology , Oxidants/toxicity , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Smad Proteins/drug effects , Smad Proteins/metabolism , Sp7 Transcription Factor/drug effects , Sp7 Transcription Factor/metabolism
20.
Medicine (Baltimore) ; 99(8): e18912, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32080074

ABSTRACT

BACKGROUND: Knee osteoarthritis (KOA) is the most common form of degenerative arthritis. We used Phellinus linteus (PL), which has been well-known anti-inflammatory function. In this study, we will evaluate if PL extract improves symptoms with KOA. METHODS: This study will be an 8-week single-center randomized controlled double-blind clinical trial. Total of 24 subjects with KOA will be enrolled and they will be divided into 3 groups, PL 1,000 mg, PL 1,500 mg and placebo. Subjects will be followed up every 4 weeks with efficacy and safety at the 2nd and 3rd visits. All subjects should maintain a dosage schedule for this protocol. The primary outcome will be assessed with the Korean version of the Western Ontario and McMasters Universities. And the secondary outcomes will be measured using the visual analog scale, quality of life scale (EQ-5D-3L), ESR, C-reactive protein, and C-telopeptide of type-II collagen. Statistical analysis will be performed on the principle of full analysis set. DISCUSSION: This study has inclusion and exclusion criteria and a well-controlled intervention. This clinical trial is the first step to assess the efficacy and safety of PL in patients with KOA. This study will make an important contribution to the literature and aid follow-up research into the use of PL in KOA.


Subject(s)
Cartilage, Articular/drug effects , Knee Joint/drug effects , Osteoarthritis, Knee/drug therapy , Plant Extracts/administration & dosage , Administration, Oral , Adult , Aged , Blood Sedimentation/drug effects , C-Reactive Protein/drug effects , Collagen Type I/drug effects , Humans , Middle Aged , Osteoarthritis, Knee/blood , Osteoarthritis, Knee/pathology , Peptides/drug effects , Phellinus , Placebos/administration & dosage , Plant Extracts/therapeutic use , Prospective Studies , Quality of Life , Republic of Korea/epidemiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...