Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Curr Obes Rep ; 12(3): 308-331, 2023 Sep.
Article En | MEDLINE | ID: mdl-37578613

PURPOSE OF REVIEW: Metabolic syndrome (MetS), also called the 'deadly quartet' comprising obesity, diabetes, dyslipidemia, and hypertension, has been ascertained to have a causal role in the pathogenesis of osteoarthritis (OA). This review is aimed at discussing the current knowledge on the contribution of metabolic syndrome and its various components to OA pathogenesis and progression. RECENT FINDINGS: Lately, an increased association identified between the various components of metabolic syndrome (obesity, diabetes, dyslipidemia, and hypertension) with OA has led to the identification of the 'metabolic phenotype' of OA. These metabolic perturbations alongside low-grade systemic inflammation have been identified to inflict detrimental effects upon multiple tissues of the joint including cartilage, bone, and synovium leading to complete joint failure in OA. Recent epidemiological and clinical findings affirm that adipokines significantly contribute to inflammation, tissue degradation, and OA pathogenesis mediated through multiple signaling pathways. OA is no longer perceived as just a 'wear and tear' disease and the involvement of the metabolic components in OA pathogenesis adds up to the complexity of the disease. Given the global surge in obesity and its allied metabolic perturbations, this review aims to throw light on the current knowledge on the pathophysiology of MetS-associated OA and the need to address MetS in the context of metabolic OA management. Better regulation of the constituent factors of MetS could be profitable in preventing MetS-associated OA. The identification of key roles for several metabolic regulators in OA pathogenesis has also opened up newer avenues in the recognition and development of novel therapeutic agents.


Dyslipidemias , Hypertension , Metabolic Syndrome , Osteoarthritis , Humans , Metabolic Syndrome/complications , Osteoarthritis/etiology , Osteoarthritis/metabolism , Obesity , Inflammation/complications , Inflammation/metabolism
2.
Cell Tissue Res ; 391(1): 127-144, 2023 Jan.
Article En | MEDLINE | ID: mdl-36227376

Obesity (Ob) depicts a state of energy imbalance(s) being characterized by the accumulation of excessive fat and which predisposes to several metabolic diseases. Mesenchymal stem cells (MSCs) represent a promising option for addressing obesity and its associated metabolic co-morbidities. The present study aims at assessing the beneficial effects of human placental MSCs (P-MSCs) in mitigating Ob-associated insulin resistance (IR) and mitochondrial dysfunction both in vivo and in vitro. Under obesogenic milieu, adipocytes showed a significant reduction in glucose uptake, and impaired insulin signaling with decreased expression of UCP1 and PGC1α, suggestive of dysregulated non-shivering thermogenesis vis-a-vis mitochondrial biogenesis respectively. Furthermore, obesogenic adipocytes demonstrated impaired mitochondrial respiration and energy homeostasis evidenced by reduced oxygen consumption rate (OCR) and blunted ATP/NAD+/NADP+ production respectively. Interestingly, co-culturing adipocytes with P-MSCs activated PI3K-Akt signaling, improved glucose uptake, diminished ROS production, enhanced mitochondrial OCR, improved ATP/NAD+/NADP+ production, and promoted beiging of adipocytes evidenced by upregulated expression of PRDM16, UCP1, and PGC1α expression. In vivo, P-MSCs administration increased the peripheral blood glucose uptake and clearance, and improved insulin sensitivity and lipid profile with a coordinated increase in the ratio of ATP/ADP and NAD+ and NADP+ in the white adipose tissue (WAT), exemplified in WNIN/GR-Ob obese mutant rats. In line with in vitro findings, there was a significant reduction in adipocyte hypertrophy, increased mitochondrial staining, and thermogenesis. Our findings advocate for a therapeutic application of P-MSCs for improving glucose and energy homeostasis, i.e., probably restoring non-shivering thermogenesis towards obesity management.


Adipocytes , Energy Metabolism , Glucose , Insulin Resistance , Mesenchymal Stem Cells , Obesity , Placenta , Animals , Female , Humans , Rats , Adenosine Triphosphate/metabolism , Adipocytes/metabolism , Glucose/metabolism , Homeostasis , Insulin Resistance/physiology , Mesenchymal Stem Cells/metabolism , NAD/metabolism , NADP/metabolism , Obesity/metabolism , Obesity/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Placenta/metabolism , Glucose Metabolism Disorders/pathology , Energy Metabolism/physiology
3.
Hum Cell ; 35(2): 557-571, 2022 Mar.
Article En | MEDLINE | ID: mdl-35091972

Obesity poses a significant risk factor for the onset of metabolic syndrome with allied complications, wherein mesenchymal stem cell therapy is seen as a promising treatment for obesity-induced metabolic syndrome. In the present study, we aim to explore the beneficial effects of the human placental mesenchymal stromal cells (P-MSCs) on obesity-associated insulin resistance (IR) including inflammation. To understand this, we have analyzed the peripheral blood glucose, serum insulin levels by ELISA, and the glucose uptake capacity of skeletal muscle by a 2-NBDG assay using flow cytometry in WNIN/GR-Ob rats treated with and without P-MSCs. Also, we have studied insulin signaling and cytokine profile in the skeletal muscle by western blotting, dot blotting, and Multiplex-ELISA techniques. The skeletal muscle of WNIN/GR-Ob rats demonstrates dysregulation of cytokines, altered glucose uptake vis-a-vis insulin signaling. However, P-MSCs' treatment was effective in WNIN/GR-Ob rats as compared to its control, to restore HOMA-IR, re-establishes dysregulated cytokines and PI3K-Akt pathway in addition to enhanced Glut4 expression and glucose uptake studied in skeletal muscle. Overall, our data advocate the beneficial effects of P-MSCs to ameliorate inflammatory milieu, improve insulin sensitivity, and normalize glucose homeostasis underlining the Ob-T2D conditions, and we attribute for immunomodulatory, paracrine, autocrine, and multipotent functions of P-MSCs.


Diabetes Mellitus, Type 2 , Mesenchymal Stem Cells , Animals , Cytokines , Female , Humans , Insulin/metabolism , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Obesity/therapy , Phosphatidylinositol 3-Kinases , Placenta , Pregnancy , Rats
5.
Inflammopharmacology ; 29(6): 1701-1717, 2021 Dec.
Article En | MEDLINE | ID: mdl-34546477

Osteoarthritis (OA) is the most prevalent joint disease predominantly characterized by inflammation which drives cartilage destruction. Mesenchymal stem cells-condition medium (MSC-CM) or the secretome is enriched with bioactive factors and possesses anti-inflammatory and regenerative effects. The present study aimed at evaluating the effects of combining MSC-conditioned medium with stigmasterol compared with the individual treatments in alleviating interleukin-1 beta (IL-1ß)-induced inflammation in rat chondrocytes. Stigmasterol is a phytosterol exhibiting anti-inflammatory effects. IL-1ß (10 ng/ml) was used to induce inflammation and mimic OA in-vitro in primary rat articular chondrocytes. The IL-1ß-stimulated chondrocytes were treated with MSC-CM, stigmasterol, and a combination of MSC-CM and stigmasterol for 24 h. Cell viability was measured using MTT assay. Protein expression of inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), collagen II (COL2A1) and matrix metalloproteinase (MMP)-13 were evaluated by immunofluorescence. Gene expression levels of MMP-3, MMP-13 and A Disintegrin-like and Metalloproteinases with Thrombospondin Motifs (ADAMTS)-5 were measured using qRT-PCR. NF-κB signaling pathway was studied using western blotting. A significant reduction in the expression of iNOS, IL-6, MMP-3, MMP-13 and ADAMTS-5, and a significant increase in COL2A1 expression was observed in the rat chondrocytes across all the treatment groups. However, the combination treatment of MSC-CM and stigmasterol remarkably reversed the IL-1ß-induced pro-inflammatory/pro-catabolic responses to near normal levels comparable to the control group. The combination treatment (MSC-CM + stigmasterol) elicited a superior anti-inflammatory/anti-catabolic effect by inhibiting the IL-1ß-induced NF-κB activation evidenced by the negligible phosphorylation of p65 and IκBα subunits, thereby emphasizing the benefit of the combination therapy over the individual treatments.


Inflammation/drug therapy , Mesenchymal Stem Cells/cytology , Osteoarthritis/drug therapy , Stigmasterol/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Chondrocytes/pathology , Combined Modality Therapy , Disease Models, Animal , Female , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Rats , Rats, Wistar , Secretome/metabolism , Stigmasterol/administration & dosage
6.
Sci Rep ; 11(1): 16983, 2021 08 20.
Article En | MEDLINE | ID: mdl-34417511

Obesity (Ob) poses a significant risk factor for the onset of metabolic syndrome with associated complications, wherein the Mesenchymal Stem Cell (MSC) therapy shows pre-clinical success. Here, we explore the therapeutic applications of human Placental MSCs (P-MSCs) to address Ob-associated Insulin Resistance (IR) and its complications. In the present study, we show that intramuscular injection of P-MSCs homed more towards the visceral site, restored HOMA-IR and glucose homeostasis in the WNIN/GR-Ob (Ob-T2D) rats. P-MSC therapy was effective in re-establishing the dysregulated cytokines. We report that the P-MSCs activates PI3K-Akt signaling and regulates the Glut4-dependant glucose uptake and its utilization in WNIN/GR-Ob (Ob-T2D) rats compared to its control. Our data reinstates P-MSC treatment's potent application to alleviate IR and restores peripheral blood glucose clearance evidenced in stromal vascular fraction (SVF) derived from white adipose tissue (WAT) of the WNIN/GR-Ob rats. Gaining insights, we show the activation of the PI3K-Akt pathway by P-MSCs both in vivo and in vitro (palmitate primed 3T3-L1 cells) to restore the insulin sensitivity dysregulated adipocytes. Our findings suggest a potent application of P-MSCs in  pre-clinical/Ob-T2D management.


Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Experimental/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Models, Biological , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/therapy , Female , Glucose Transporter Type 4/metabolism , Homeostasis , Humans , Insulin/metabolism , Macrophages/metabolism , Obesity/complications , Phosphatidylinositol 3-Kinases/metabolism , Placenta/cytology , Pregnancy , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction
7.
Stem Cell Investig ; 8: 5, 2021.
Article En | MEDLINE | ID: mdl-33829057

Mesenchymal stem cells (MSCs) have gained wide therapeutic acceptance in regenerative medicine due to their potential in repair process in restoring the damaged tissues and controlling inflammation. In the present study, we report for the first time the beneficial effects of combining placental-derived MSCs (hPMSCs) with stigmasterol-a plant-derived sterol to accelerate cartilage repair and regeneration in a monosodium-iodoacetate (MIA) induced osteoarthritis (OA) rat model. Control animals (Group I) received no treatment. Experimental animals (Group II) received a single intra-articular injection of MIA (2 mg) in the right knee joints. The Group II animals developed OA-like lesions within a week of MIA injection. They were subdivided further as: (II-A): OA, (II-B): OA+hPMSCs (2×106 cells, single-dose/intra-articular injection), (II-C): OA+stigmasterol (20 µg/mL, single-dose/intra-articular injection) and (II-D): OA+hPMSCs+stigmasterol. The animals were monitored for four more weeks after which they were sacrificed, the right limbs dissected out and assessed for cartilage repair and regeneration using micro-computed tomography (micro-CT) and histology. Results showed that the combined administration of hPMSCs with stigmasterol (II-D) was the most effective in correcting the OA lesions, with concomitant repair and regeneration. However, hPMSCs (II-B) or stigmasterol (II-C) per se treated groups showed only marginal beneficial effects and were not significant. Thus the present study provides valuable insights in situ using a combination of hPMSCs and stigmasterol towards cartilage repair and regeneration. We advocate the participation of populating cells or residual chondrocytes in addition to its anti-inflammatory functions.

...