10 research outputs found

    Sex Hormone Binding Globulin (SHBG) Mitigates ER Stress in Hepatocytes In Vitro and Ex Vivo

    No full text
    Despite multiple research studies regarding metabolic syndrome and diabetes, the full picture of their molecular background and pathogenies remains elusive. The latest studies revealed that sex hormone-binding globulin (SHBG)—a serum protein released mainly by the liver—may participate in metabolic dysregulation, as its low serum level correlates with a risk for obesity, metabolic syndrome, and diabetes. Yet, the molecular phenomenon linking SHBG with these disorders remains unclear. In the presented study, we investigate how exogenous SHBG affects metabolically impaired hepatocytes with special attention to endoplasmic reticulum stress (ER stress) and lipid metabolism both in vitro and ex vivo. For that reason, palmitate-treated HepG2 cells and liver tissue samples collected post mortem were cultured in the presence of 50 nM and 100 nM SHBG. We found that SHBG protects against ER stress development and its progression. We have found that SHBG decreased the expression levels of inositol-requiring enzyme 1 (IRE1α), activating transcription factor 6 (ATF6), DNA damage-inducible transcript 3 (CHOP), and immunoglobulin heavy chain-binding protein (BIP). Furthermore, we have shown that it regulates lipolytic gene expression ex vivo. Additionally, herein, we deliver a novel large-animal model to study SHBG in translational research. Our data provide new insights into the cellular and molecular mechanisms by which SHBG modulates hepatocyte metabolism and offer a new experimental approach to study SHBG in human diseases

    Inhibition of the Low Molecular Weight Protein Tyrosine Phosphatase (LMPTP) as a Potential Therapeutic Strategy for Hepatic Progenitor Cells Lipotoxicity—Short Communication

    No full text
    Equine metabolic syndrome (EMS) is a cluster of metabolic disorders, such as obesity, hyperinsulinemia, and hyperleptinemia, as well as insulin resistance (IR). In accordance with the theory linking obesity and IR, excessive accumulation of lipids in insulin-sensitive tissues (lipotoxicity), like liver, alters several cellular functions, including insulin signaling. Therefore, the purpose of the study was to isolate equine hepatic progenitor-like cells (HPCs) and assess whether inhibition of low molecular weight protein tyrosine phosphatase (LMPTP) affects the expression of genes involved in macroautophagy, chaperone-mediated autophagy (CMA), endoplasmic reticulum stress, and mitochondrial dynamics in a palmitate-induced IR model. We demonstrated that LMPTP inhibition significantly enhanced expression of heat shock cognate 70 kDa protein (HSC70), lysosome-associated membrane protein 2 (LAMP2), and parkin (PRKN), all master regulators of selective autophagy. We also observed downregulation of C/EBP homologous protein (CHOP), activating transcription factor 6 (ATF6) and binding immunoglobulin protein encoded by the HSPA gene. Moreover, LMPTP inhibition increased alternative splicing of X-box binding protein 1 (XBP1), suggesting high endonuclease activity of inositol-requiring enzyme 1 alpha (IRE1α). Taken together, our data provide convincing evidence that LMPTP inhibition reverses palmitate-induced insulin resistance and lipotoxicity. In conclusion, this study highlights the role of LMPTP in the regulation of CMA, mitophagy, and ER stress, and provides a new in vitro model for studying HPC lipotoxicity in pre-clinical research

    The Hepatic Stellate Cells (HSTCs) and Adipose-derived Mesenchymal Stem Cells (ASCs) Axis as a Potential Major Driver of Metabolic Syndrome – Novel Concept and Therapeutic Implications

    No full text
    Herein, we would like to introduce a novel concept for the prevention and treatment of metabolic syndrome, which is based on molecular relationship between liver and adipose tissue. Particularly, we believe, that unravelling the molecular crosstalk between hepatokines and adipokines will allow to better understand the pathophysiology of metabolic diseases and allow to develop novel, effective therapeutic solutions against obesity and metabolic syndrome. Inter-organ communication on the level of stem progenitor cells-hepatic stellate cells (HSTCs) and adipose-derived progenitors (ASCs) could represents a key mechanism involved in controlling glucose tolerance as well as insulin sensitivity

    <p>Fe<sub>3</sub>O<sub>4</sub> Magnetic Nanoparticles Under Static Magnetic Field Improve Osteogenesis via RUNX-2 and Inhibit Osteoclastogenesis by the Induction of Apoptosis</p>

    No full text
    International audiencePurpose: The presented study aimed to investigate the effects of Fe 3O 4 nanoparticles and static magnetic field on osteoblast and osteoclasts’ metabolic activity.Methods: Magnetic nanoparticles were prepared by a wet chemical co-precipitation process and analyzed using X-ray powder diffraction, high-resolution transmission electron microscope (HRTEM), dynamic light scattering (DLS), laser Doppler velocimetry, Raman and the Mössbauer spectroscopy. In vitro experiments were performed using MC3T3, 4B12 and RAW 264.7 cell lines. Cells were cultured in the presence of nanoparticles and with or without exposure to the magnetic field. Proteins were investigated with Western blotting and immunofluorescence and Western blot. Gene expression was analyzed with a quantitative real-time polymerase chain reaction.Results: Obtained particles were in the nano-range (average size around 50 nm) and had a spherical-like morphology. The typical hydrodynamic size was in the range 178– 202 nm and Zeta potential equaled – 9.51 mV. Mössbauer spectrum corresponds to the Fe+3 ions in tetrahedral (A) and Fe+3 and Fe+2 ions in octahedral (B) sites of Fe 3O 4. In vitro study revealed cytocompatibility and anti-inflammatory effects of fabricated nanoparticles. Furthermore, it was shown that nanoparticles combined with magnetic field exposure enhance osteogenic differentiation of MC3T3 cells by upregulation of RUNX-2 activity. Under the same experimental condition, nanoparticles and magnetic field decreased osteoclastogenesis of 4B12 by the induction of apoptosis through the mitochondrial-dependent pathway.Conclusion: Fe 3O 4 nanoparticles together with magnetic field can be applied for the fabrication of novel biomaterials for the treatment of bone disorders related to bone loss in which a balance between bone-forming and resorbing cells is disturb

    Aminopropyltriethoxysilane (APTES)-Modified Nanohydroxyapatite (nHAp) Incorporated with Iron Oxide (IO) Nanoparticles Promotes Early Osteogenesis, Reduces Inflammation and Inhibits Osteoclast Activity

    No full text
    Due to its increased prevalence, osteoporosis (OP) represents a great challenge to health care systems and brings an economic burden. To overcome these issues, treatment plans that suit the need of patients should be developed. One of the approaches focuses on the fabrication of personalized biomaterials, which can restore the balance and homeostasis of disease-affected bone. In the presented study, we fabricated nanometer crystalline hydroxyapatite (nHAp) and iron oxide (IO) nanoparticles stabilized with APTES and investigated whether they can modulate bone cell metabolism and be useful in the fabrication of personalized materials for OP patients. Using a wide range of molecular techniques, we have shown that obtained nHAp@APTES promotes viability and RUNX-2 expression in osteoblasts, as well as reducing activity of critical proinflammatory cytokines while inhibiting osteoclast activity. Materials with APTES modified with nHAp incorporated with IO nanoparticles can be applied to support the healing of osteoporotic bone fractures as they enhance metabolic activity of osteoblasts and diminish osteoclasts&rsquo; metabolism and inflammation

    Novel Nanohydroxyapatite (nHAp)-Based Scaffold Doped with Iron Oxide Nanoparticles (IO), Functionalized with Small Non-Coding RNA (miR-21/124) Modulates Expression of Runt-Related Transcriptional Factor 2 and Osteopontin, Promoting Regeneration of Osteoporotic Bone in Bilateral Cranial Defects in a Senescence-Accelerated Mouse Model (SAM/P6). PART 2

    No full text
    International audiencePurpose: Healing of osteoporotic defects is challenging and requires innovative approaches to elicit molecular mechanisms promoting osteoblasts-osteoclasts coupling and bone homeostasis.Methods: Cytocompatibility and biocompatibility of previously characterised nanocomposites, i.e Ca5(PO4)3OH/Fe3O4 (later called nHAp/IO) functionalised with microRNAs (nHAp/IO@miR-21/124) was tested. In vitro studies were performed using a direct co-culture system of MC3T3-E1 pre-osteoblast and 4B12 pre-osteoclasts. The analysis included determination of nanocomposite influence on cultures morphology (confocal imaging), viability and metabolic activity (Alamar Blue assay). Pro-osteogenic signals were identified at mRNA, miRNA and protein level with RT-qPCR, Western blotting and immunocytochemistry. Biocompatibility of biomaterials was tested using bilateral cranial defect performed on a senescence-accelerated mouse model, ie SAM/P6 and Balb/c. The effect of biomaterial on the process of bone healing was monitored using microcomputed tomography.Results: The nanocomposites promoted survival and metabolism of bone cells, as well as enhanced functional differentiation of pre-osteoblasts MC3T3-E1 in co-cultures with pre-osteoclasts. Differentiation of MC3T3-E1 driven by nHAp/IO@miR-21/124 nanocomposite was manifested by improved extracellular matrix differentiation and up-regulation of pro-osteogenic transcripts, ie late osteogenesis markers. The nanocomposite triggered bone healing in a cranial defect model in SAM/P6 mice and was replaced by functional bone in Balb/c mice.Conclusion: This study demonstrates that the novel nanocomposite nHAp/IO can serve as a platform for therapeutic miRNA delivery. Obtained nanocomposite elicit pro-osteogenic signals, decreasing osteoclasts differentiation, simultaneously improving osteoblasts metabolism and their transition toward pre-osteocytes and bone mineralisation. The proposed scaffold can be an effective interface for in situ regeneration of osteoporotic bone, especially in elderly patients
    corecore