Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Bio Protoc ; 13(22): e4885, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38023794

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) hold immense promise in regenerative medicine as they can differentiate into various cell lineages, including adipocytes, osteoblasts, and chondrocytes. Precisely guiding hiPSC-derived mesenchymal progenitor cells (iMSCs) towards specific differentiation pathways is crucial for harnessing their therapeutic potential in tissue engineering, disease modeling, and regenerative therapies. To achieve this, we present a comprehensive and reproducible protocol for effectively differentiating iMSCs into adipocytes and osteoblasts. The differentiation process entails culturing iMSCs in tailored media supplemented with specific growth factors, which act as cues to initiate adipogenic or osteogenic commitment. Our protocol provides step-by-step guidelines for achieving adipocyte and osteoblast differentiation, ensuring the generation of mature and functional cells. To validate the success of differentiation, key assessment criteria are employed. For adipogenesis, the presence of characteristic lipid droplets within the iMSC-derived cells is considered indicative of successful differentiation. Meanwhile, Alizarin Red staining serves as a marker for the osteogenic differentiation, confirming the formation of mineralized nodules. Importantly, the described method stands out due to its simplicity, eliminating the need for specialized equipment, expensive materials, or complex reagents. Its ease of implementation offers an attractive advantage for researchers seeking robust and cost-effective approaches to derive adipocytes and osteoblasts from iMSCs. Overall, this protocol establishes a foundation for exploring the therapeutic potential of hiPSC-derived cells and advancing the field of regenerative medicine. Key features • iMSC derivation in this protocol uses embryonic body formation technique. • Adipogenesis and osteogenesis protocols were optimized for human iPSC-derived iMSCs. • Derivation of iMSC from hiPSC was developed in a feeder-free culture condition. • This protocol does not include human iPSC reprogramming strategies.

2.
Biomed Pharmacother ; 96: 198-207, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28987943

ABSTRACT

BACKGROUND/OBJECTIVE: Osteoarthritis (OA) is a leading cause of joint dysfunction, disability and poor quality of life in the affected population. The underlying mechanism of joint dysfunction involves increased oxidative stress, inflammation, high levels of cartilage extracellular matrix degrading proteases and decline in autophagy-a mechanism of cellular defense. There is no disease modifying therapies currently available for OA. Different parts of the Butea monosperma (Lam.) plant have widely been used in the traditional Indian Ayurvedic medicine system for the treatment of various human diseases including inflammatory conditions. Here we studied the chondroprotective effect of hydromethanolic extract of Butea monosperma (Lam.) flowers (BME) standardized to the concentration of Butein on human OA chondrocytes stimulated with IL-1ß. METHODS: The hydromethanolic extract of Butea monosperma (Lam.) (BME) was prepared with 70% methanol-water mixer using Soxhlet. Chondrocytes viability after BME treatment was measured by MTT assay. Gene expression levels were determined by quantitative polymerase chain reaction (qPCR) using TaqMan assays and immunoblotting with specific antibodies. Autophagy activation was determined by measuring the levels of microtubule associated protein 1 light chain 3-II (LC3-II) by immunoblotting and visualization of autophagosomes by transmission electron and confocal microscopy. RESULTS: BME was non-toxic to the OA chondrocytes at the doses employed and suppressed the IL-1ß induced expression of inerleukin-6 (IL-6) and matrix metalloprotease-3 (MMP-3), MMP-9 and MMP-13. BME enhanced autophagy in chondrocytes as determined by measuring the levels of LC3-II by immunoblotting and increased number of autophagosomes in BME treated chondrocytes by transmission electron microscopy and confocal microscopy. BME upregulated the expression of several autophagy related genes and increased the autophagy flux in human OA chondrocytes under pathological conditions. Further analysis revealed that BME activated autophagy in chondrocytes via inhibition of mammalian target of rapamycin (mTOR) pathway. Of importance is our finding that BME-mediated suppression of IL-1ß induced expression of IL-6, MMP-3, -9, and -13 was autophagy dependent and was abrogated by inhibition of autophagy. CONCLUSION: The above results show that the Butea monosperma (Lam.) extract has strong potential to activate autophagy and suppress IL-1ß induced expression of IL-6 and MMP-3, -9 and -13 in human OA chondrocytes. This study shows that BME or compounds derived from BME can be developed as safe and effective chondroprotective agent(s) that function by activating autophagy to suppress the expression of inflammatory and catabolic factors associated with OA pathogenesis.


Subject(s)
Butea , Chondrocytes/metabolism , Interleukin-1beta/pharmacology , Interleukin-6/biosynthesis , Matrix Metalloproteinases/biosynthesis , Osteoarthritis/metabolism , Aged , Autophagy/drug effects , Autophagy/physiology , Chondrocytes/drug effects , Dose-Response Relationship, Drug , Flowers , Gene Expression , Humans , Interleukin-6/genetics , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 3/biosynthesis , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/genetics , Middle Aged , Plant Extracts/isolation & purification , Plant Extracts/pharmacology
3.
Phytother Res ; 31(5): 778-782, 2017 May.
Article in English | MEDLINE | ID: mdl-28276100

ABSTRACT

Pomegranate fruit extract (PE) rich in polyphenols has been shown to exert chondroprotective effects, but the mechanism is not established. Here, we used an in vitro model of inflammation in osteoarthritis (OA) to investigate the potential of PE to suppress interleukin 1 beta (IL-1ß)-stimulated expression of inflammatory cytokine IL-6, generation of reactive oxygen species (ROS) levels, and investigated the mechanism of NF-κB inhibition by analyzing the activation of the kinases upstream of IκBα in primary human chondrocytes. Total and phosphorylated forms of kinases and expression of IL-6 were determined at protein and mRNA levels by western immunoblotting and Taqman assay, respectively. Dihydrorhodamine 123 staining estimated ROS generation. Pomegranate fruit extract inhibited the mRNA and protein expression of IL-6, generation of ROS, and inhibited the IL-1ß-mediated phosphorylation of inhibitor of nuclear factor kappa-B kinase subunit beta (IKKß), expression of IKKß mRNA, degradation of IκBα, and activation and nuclear translocation of NF-κB/p65 in human chondrocytes. Importantly, phosphorylation of NF-κB-inducing kinase was blocked by PE in IL-1ß-treated human OA chondrocytes. Taken together, these data suggest that PE exerts the chondroprotective effect(s) by suppressing the production of IL-6 and ROS levels. Inhibition of NF-κB activation by PE was blocked via modulation of activation of upstream kinases in human OA chondrocytes. Copyright © 2017 John Wiley & Sons, Ltd.


Subject(s)
I-kappa B Kinase/metabolism , Interleukin-6/metabolism , Lythraceae/chemistry , NF-kappa B/metabolism , Plant Extracts/pharmacology , Protein Serine-Threonine Kinases/metabolism , Chondrocytes/drug effects , Fruit/chemistry , Gene Expression Regulation/drug effects , Humans , I-kappa B Kinase/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , NF-kappa B/genetics , Phosphorylation/drug effects , Plant Extracts/chemistry , Polyphenols/pharmacology , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , NF-kappaB-Inducing Kinase
4.
Sci Rep ; 7: 43789, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256567

ABSTRACT

Osteoarthritis (OA) is a common joint disorder with varying degrees of inflammation and sustained oxidative stress. The root extract of Scutellaria baicalensis (SBE) has been used for the treatment of inflammatory and other diseases. Here, we performed activity-guided HPLC-fractionation of SBE, identified the active ingredient(s) and investigated its chondroprotective potential. We found that the Wogonin containing fraction-4 (F4) was the most potent fraction based on its ability to inhibit ROS production and the suppression of catabolic markers including IL-6, COX-2, iNOS, MMP-3, MMP-9, MMP-13 and ADAMTS-4 in IL-1ß-treated OA chondrocytes. OA chondrocytes treated with F4 in the presence of IL-1ß showed significantly enhanced expression of anabolic genes ACAN and COL2A1. In an in vitro model of cartilage degradation treatment with F4 inhibited s-GAG release from IL-1ß-treated human cartilage explants. The inhibitory effect of F4 was not mediated through the inhibition of MAPKs and NF-κB activation but was mediated through the suppression of c-Fos/AP-1 activity at transcriptional and post transcriptional levels in OA chondrocytes. Purified Wogonin mimicked the effects of F4 in IL-1ß-stimulated OA chondrocytes. Our data demonstrates that a Wogonin-rich fraction of SBE exert chondroprotective effects through the suppression of c-Fos/AP-1 expression and activity in OA chondrocytes under pathological conditions.


Subject(s)
Chondrocytes/drug effects , Flavanones/pharmacology , Plant Extracts/pharmacology , Plant Roots/chemistry , Scutellaria baicalensis/chemistry , Transcription Factor AP-1/metabolism , Cells, Cultured , Chemical Fractionation/methods , Chondrocytes/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Osteoarthritis/pathology , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/genetics
5.
Nutrition ; 33: 1-13, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27908544

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is characterized by cartilage degradation in the affected joints. Pomegranate fruit extract (PFE) inhibits cartilage degradation in vitro. The aim of this study was to determine whether oral consumption of PFE inhibits disease progression in rabbits with surgically induced OA. METHODS: OA was surgically induced in the tibiofemoral joints of adult New Zealand White rabbits. In one group, animals were fed PFE in water for 8 wk postsurgery. In the second group, animals were fed PFE for 2 wk before surgery and for 8 wk postsurgery. Histologic assessment and scoring of the cartilage was per Osteoarthritis Research Society International guidelines. Gene expression and matrix metalloproteinases (MMP) activity were determined using quantitative reverse transcriptase polymerase chain reaction and fluorometric assay, respectively. Interleukin (IL)-1 ß, MMP-13, IL-6, prostaglandin (PG)E2, and type II collagen (COL2A1) levels in synovial fluid/plasma/culture media were quantified using enzyme-linked immunosorbent assay. Expression of active caspase-3 and poly (ADP-ribose) polymerase p85 was determined by immunohistochemistry. Effect of PFE and inhibitors of MMP-13, mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB was studied in IL-1 ß-stimulated rabbit articular chondrocytes. RESULTS: Safranin-O-staining and chondrocyte cluster formation was significantly reduced in the anterior cruciate ligament transaction plus PFE fed groups. Expression of MMP-3, MMP-9, and MMP-13 mRNA was higher in the cartilage of rabbits given water alone but was significantly lower in the animals fed PFE. PFE-fed rabbits had lower IL-6, MMP-13, and PGE2 levels in the synovial fluid and plasma, respectively, and showed higher expression of aggrecan and COL2A1 mRNA. Significantly higher numbers of chondrocytes were positive for markers of apoptosis in the joints of rabbits with OA given water only compared with those in the PFE-fed groups. PFE pretreatment significantly reduced IL-1 ß induced IL-6 and MMPs expression in rabbit articular chondrocytes. These effects were also mimicked using MMP-13, MAPK, and NF-κB inhibitors in IL-1 ß-stimulated rabbit chondrocytes. In an in vitro activity assay, PFE blocked the activity of MMP-13. Like MAPK and NF-κB inhibitors, PFE was also effective in inhibiting IL-1 ß-induced PGE2 production in rabbit chondrocytes. PFE also reversed the inhibitory effect of IL-1ß on COL2A1 mRNA and protein expression in IL-1 ß-stimulated rabbit chondrocytes. CONCLUSION: The present data highlight the chondroprotective effects of PFE oral consumption in a model of posttraumatic OA and suggest that PFE-derived compounds may have potential value in the management of OA.


Subject(s)
Cartilage/drug effects , Dinoprostone/metabolism , Joints/drug effects , Lythraceae , Metalloproteases/metabolism , Osteoarthritis/drug therapy , Phytotherapy , Animals , Anterior Cruciate Ligament/drug effects , Anterior Cruciate Ligament/metabolism , Anterior Cruciate Ligament/pathology , Apoptosis , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/genetics , Collagen Type II/metabolism , Disease Models, Animal , Disease Progression , Female , Fruit , Interleukins/metabolism , Joints/cytology , Joints/metabolism , Joints/pathology , Male , Metalloproteases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , RNA, Messenger/metabolism , Rabbits , Synovial Fluid/metabolism
6.
CNS Neurol Disord Drug Targets ; 13(8): 1440-57, 2014.
Article in English | MEDLINE | ID: mdl-25345504

ABSTRACT

The increasing worldwide prevalence of type 2 diabetes mellitus (T2DM) and associated neurological disorders (NDs), such as Alzheimer disease and Parkinson's disease, have raised concerns about increasing health care and financial burden. Due to the overwhelming growth rate of T2DM and its strong association with NDs, there is an ever-growing and an urgent need to improve the diagnosis and management of the disease. Major hurdles in the management of T2DM comprise of striving for glycemic targets, polypharmacy, patient adherence and clinical inertia. The challenges occurring in the treatment of T2DM are mainly attributed to the complex heterogeneous nature of the disease and its close association with a wide variety of neurological, metabolic and cardiovascular disorders. To overcome these challenges, authors propose to focus on the treatment strategies that employ shared pathogenesis and common molecular denominators involved in the aetiology of T2DM and associated NDs. Impaired insulin signalling (as a result of perturbed redox status), insulin resistance and mitochondrial dysfunction are key molecular events that may lead to the pathogenesis of T2DM and associated NDs. However, effective management of these therapeutic strategies requires holistic experimental evidence from animal as well as clinical human studies. Therefore, a shift in the treatment paradigm from single point glycemic control to shared pathogenesis control would be an ideal approach to combat the alarming progression of diabetes and associated NDs. Therapeutic interventions focused on shared molecular pathogenesis, along with effective glycemic control, may provide protection from associated NDs.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Disease Management , Nervous System Diseases/therapy , Animals , Blood Glucose , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/etiology , Humans , Insulin Resistance , Nervous System Diseases/complications , Nervous System Diseases/epidemiology , Nervous System Diseases/etiology , Signal Transduction/physiology
7.
J Med Microbiol ; 63(Pt 3): 355-366, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24307637

ABSTRACT

Antioxidants are known to exhibit numerous health benefits including anti-ageing, anti-apoptotic and immuno-stimulatory effects. However, we present the data showing counterproductive effects of therapeutically relevant antioxidants on bacterial clearance by the immune system in a murine peritonitic model. The antioxidants ascorbic acid, glutathione and N-acetylcysteine augmented morbidity and mortality in mice carrying Eshcerichia coli-induced acute bacterial peritonitis. Treatment of peritonitic mice with antioxidants significantly increased their bacterial load in the range of 0.3-2 logs. Antioxidant administration to peritonitic mice resulted in decreased numbers of macrophages, B-cells and dendritic cells at the primary site of infection and increased neutrophil infiltration. Serum TNF-α levels were also decreased in antioxidant-treated peritonitic mice. In vitro experiments showed that antioxidants reduced the phagocytic efficacy of peritoneal macrophages by ~60-75% and also decreased E. coli-induced oxidative burst in macrophages cells. Taken together, our data indicate that the antioxidants increased the severity of peritonitis by decreasing the phagocytic efficiency, oxidative burst, and TNF-α production, and increasing neutrophil infiltration. Based on these results, we propose that antioxidant supplementation during the course of bacterial infection is not recommended as it could be detrimental for the host. In addition, the present study underlines the importance of timing and context of antioxidant administration rather than indiscriminate usage to gain the best possible therapeutic advantage of these redox compounds.


Subject(s)
Antioxidants/pharmacology , Escherichia coli/immunology , Macrophages, Peritoneal/drug effects , Peritonitis/immunology , Phagocytosis/drug effects , Animals , Antioxidants/therapeutic use , Disease Models, Animal , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Escherichia coli Infections/drug therapy , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli K12/drug effects , Escherichia coli K12/immunology , Female , Macrophages, Peritoneal/metabolism , Male , Mice , Neutrophil Infiltration/drug effects , Peritonitis/drug therapy , Peritonitis/microbiology , Respiratory Burst/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/drug effects
8.
Free Radic Res ; 45(8): 975-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21651451

ABSTRACT

Recent investigations suggest that cellular redox status may play a key role in the regulation of several immune functions. Treatment of lymphocytes with vitamin K3 (menadione) resulted in a significant decrease in cellular GSH/GSSG ratio and concomitant increase in the ROS levels. It also suppressed Concanavalin A (Con A)-induced proliferation and cytokine production in lymphocytes and CD4 + T cells in vitro. Immunosuppressive effects of menadione were abrogated only by thiol containing antioxidants. Mass spectrometric analysis showed that menadione directly interacted with thiol antioxidant GSH. Menadione completely suppressed Con A-induced activation of ERK, JNK and NF-κB in lymphocytes. It also significantly decreased the homeostasis driven proliferation of syngeneic CD4 + T cells. Further, menadione significantly delayed graft-vs-host disease morbidity and mortality in mice. Menadione suppressed phytohemagglutinin-induced cytokine production in human peripheral blood mononuclear cells. These results reveal that cellular redox perturbation by menadione is responsible for significant suppression of lymphocyte responses.


Subject(s)
Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocytes/drug effects , Lymphocytes/immunology , Vitamin K 3/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Inflammation/drug therapy , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Oxidation-Reduction/drug effects , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL