Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Int J Mol Sci ; 24(9)2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37175783

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a disease characterized by a prolonged hyperglycemic condition caused by insulin resistance mechanisms in muscle and liver, reduced insulin production by pancreatic ß cells, and a chronic inflammatory state with increased levels of the pro-inflammatory marker semaphorin 3E. Phytochemicals present in several foods have been used to complement oral hypoglycemic drugs for the management of T2DM. Notably, dipeptidyl peptidase IV (DPPIV) inhibitors have demonstrated efficacy in the treatment of T2DM. Our study aimed to investigate, in in vitro models of insulin resistance, the ability of the flavanones naringenin and hesperetin, used alone and in combination with the anti-inflammatory natural molecules curcumin, polydatin, and quercetin, to counteract the insulin resistance and pro-inflammatory molecular mechanisms that are involved in T2DM development. Our results show for the first time that the combination of naringenin, hesperetin, curcumin, polydatin, and quercetin (that mirror the nutraceutical formulation GliceFen®, Mivell, Italy) synergistically decreases expression levels of the pro-inflammatory gene SEMA3E in insulin-resistant HepG2 cells and synergistically decreases DPPIV activity in insulin-resistant Hep3B cells, indicating that the combination of these five phytochemicals is able to inhibit pro-inflammatory and insulin resistance molecular mechanisms and could represent an effective innovative complementary approach to T2DM pharmacological treatment.


Subject(s)
Curcumin , Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Flavanones , Insulin Resistance , Semaphorins , Humans , Curcumin/pharmacology , Curcumin/therapeutic use , Diabetes Mellitus, Type 2/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Flavanones/chemistry , Insulin/therapeutic use , Quercetin/chemistry , Semaphorins/therapeutic use
2.
Int J Mol Sci ; 24(10)2023 May 16.
Article in English | MEDLINE | ID: mdl-37240169

ABSTRACT

During aging, bone marrow mesenchymal stromal cells (MSCs)-the precursors of osteoblasts-undergo cellular senescence, losing their osteogenic potential and acquiring a pro-inflammatory secretory phenotype. These dysfunctions cause bone loss and lead to osteoporosis. Prevention and intervention at an early stage of bone loss are important, and naturally active compounds could represent a valid help in addition to diet. Here, we tested the hypothesis that the combination of two pro-osteogenic factors, namely orthosilicic acid (OA) and vitamin K2 (VK2), and three other anti-inflammatory compounds, namely curcumin (CUR), polydatin (PD) and quercetin (QCT)-that mirror the nutraceutical BlastiMin Complex® (Mivell, Italy)-would be effective in promoting MSC osteogenesis, even of replicative senescent cells (sMSCs), and inhibiting their pro-inflammatory phenotype in vitro. Results showed that when used at non-cytotoxic doses, (i) the association of OA and VK2 promoted MSC differentiation into osteoblasts, even when cultured without other pro-differentiating factors; and (ii) CUR, PD and QCT exerted an anti-inflammatory effect on sMSCs, and also synergized with OA and VK2 in promoting the expression of the pivotal osteogenic marker ALP in these cells. Overall, these data suggest a potential role of using a combination of all of these natural compounds as a supplement to prevent or control the progression of age-related osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Curcumin , Mesenchymal Stem Cells , Osteoporosis , Humans , Osteogenesis , Quercetin/therapeutic use , Vitamin K 2/pharmacology , Vitamin K 2/metabolism , Curcumin/pharmacology , Bone Marrow/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Bone Diseases, Metabolic/metabolism , Cells, Cultured , Bone Marrow Cells
3.
Int J Mol Sci ; 24(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36982232

ABSTRACT

Sinusoidal endothelial cells are the predominant vascular surface of the bone marrow and constitute the functional hematopoietic niche where hematopoietic stem and progenitor cells receive cues for self-renewal, survival, and differentiation. In the bone marrow hematopoietic niche, the oxygen tension is usually very low, and this condition affects stem and progenitor cell proliferation and differentiation and other important functions of this region. Here, we have investigated in vitro the response of endothelial cells to a marked decrease in O2 partial pressure to understand how the basal gene expression of some relevant biological factors (i.e., chemokines and interleukins) that are fundamental for the intercellular communication could change in anoxic conditions. Interestingly, mRNA levels of CXCL3, CXCL5, and IL-34 genes are upregulated after anoxia exposure but become downmodulated by sirtuin 6 (SIRT6) overexpression. Indeed, the expression levels of some other genes (such as Leukemia Inhibitory Factor (LIF)) that were not significantly affected by 8 h anoxia exposure become upregulated in the presence of SIRT6. Therefore, SIRT6 mediates also the endothelial cellular response through the modulation of selected genes in an extreme hypoxic condition.


Subject(s)
Hematopoietic Stem Cells , Sirtuins , Hematopoietic Stem Cells/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Bone Marrow/metabolism , Interleukins/metabolism , Sirtuins/genetics , Sirtuins/metabolism
4.
Int J Mol Sci ; 24(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37047732

ABSTRACT

Sirtuin 6 (SIRT6) is a member of the mammalian NAD+-dependent deac(et)ylase sirtuin family. SIRT6's anti-inflammatory roles are emerging increasingly often in different diseases and cell types, including endothelial cells. In this study, the role of SIRT6 in pro-inflammatory conditions was investigated by engineering human umbilical vein endothelial cells to overexpress SIRT6 (SIRT6+ HUVECs). Our results showed that SIRT6 overexpression affected the levels of adhesion molecules and sustained megakaryocyte proliferation and proplatelet formation. Interestingly, the pro-inflammatory activation of the ATP/purinergic axis was reduced in SIRT6+ HUVECs. Specifically, the TNFα-induced release of ATP in the extracellular space and the increase in pannexin-1 hemichannel expression, which mediates ATP efflux, were hampered in SIRT6+ cells. Instead, NAD+ release and Connexin43 expression were not modified by SIRT6 levels. Moreover, the Ca2+ influx in response to ATP and the expression of the purinergic receptor P2X7 were decreased in SIRT6+ HUVECs. Contrary to extracellular ATP, extracellular NAD+ did not evoke pro-inflammatory responses in HUVECs. Instead, NAD+ administration reduced endothelial cell proliferation and motility and counteracted the TNFα-induced angiogenesis. Altogether, our data reinforce the view of SIRT6 activation as an anti-inflammatory approach in vascular endothelium.


Subject(s)
Human Umbilical Vein Endothelial Cells , Sirtuins , Humans , Adenosine Triphosphate , Human Umbilical Vein Endothelial Cells/metabolism , NAD , Sirtuins/metabolism , Tumor Necrosis Factor-alpha/pharmacology
5.
Molecules ; 28(19)2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37836603

ABSTRACT

The therapeutic advantages of some platinum complexes as major anticancer chemotherapeutic agents and of nucleoside analogue-based compounds as essential antiviral/antitumor drugs are widely recognized. Red blood cells (RBCs) offer a potential new strategy for the targeted release of therapeutic agents due to their biocompatibility, which can protect loaded drugs from inactivation in the blood, thus improving biodistribution. In this study, we evaluated the feasibility of loading model nucleobase-containing Pt(II) complexes into human RBCs that were highly stabilized by four N-donors and susceptible to further modification for possible antitumor/antiviral applications. Specifically, platinum-based nucleoside derivatives [PtII(dien)(N7-Guo)]2+, [PtII(dien)(N7-dGuo)]2+, and [PtII(dien)(N7-dGTP)] (dien = diethylenetriamine; Guo = guanosine; dGuo = 2'-deoxy-guanosine; dGTP = 5'-(2'-deoxy)-guanosine-triphosphate) were investigated. These Pt(II) complexes were demonstrated to be stable species suitable for incorporation into RBCs. This result opens avenues for the possible incorporation of other metalated nucleobases analogues, with potential antitumor and/or antiviral activity, into RBCs.


Subject(s)
Antineoplastic Agents , Organoplatinum Compounds , Humans , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/metabolism , Tissue Distribution , Platinum , Antineoplastic Agents/pharmacology , Antineoplastic Agents/metabolism , Antiviral Agents/pharmacology , Erythrocytes/metabolism , Guanosine/metabolism
6.
Blood ; 128(25): 2949-2959, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27733356

ABSTRACT

To begin to understand the mechanisms that regulate self-renewal, differentiation, and transformation of human hematopoietic stem cells or to evaluate the efficacy of novel treatment modalities, stem cells need to be studied in their own species-specific microenvironment. By implanting ceramic scaffolds coated with human mesenchymal stromal cells into immune-deficient mice, we were able to mimic the human bone marrow niche. Thus, we have established a human leukemia xenograft mouse model in which a large cohort of patient samples successfully engrafted, which covered all of the important genetic and risk subgroups. We found that by providing a humanized environment, stem cell self-renewal properties were better maintained as determined by serial transplantation assays and genome-wide transcriptome studies, and less clonal drift was observed as determined by exome sequencing. The human leukemia xenograft mouse models that we have established here will serve as an excellent resource for future studies aimed at exploring novel therapeutic approaches.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Stem Cell Niche , Tissue Scaffolds/chemistry , Xenograft Model Antitumor Assays , Animals , Cell Self Renewal , Cell Separation , Clone Cells , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Mesenchymal Stem Cells/cytology , Mice , Phenotype , Stromal Cells/pathology
7.
Blood ; 128(20): 2435-2449, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27574188

ABSTRACT

Deep venous thrombosis (DVT) is one of the most common cardiovascular diseases, but its pathophysiology remains incompletely understood. Although sterile inflammation has recently been shown to boost coagulation during DVT, the underlying molecular mechanisms are not fully resolved, which could potentially identify new anti-inflammatory approaches to prophylaxis and therapy of DVT. Using a mouse model of venous thrombosis induced by flow reduction in the vena cava inferior, we identified blood-derived high-mobility group box 1 protein (HMGB1), a prototypical mediator of sterile inflammation, to be a master regulator of the prothrombotic cascade involving platelets and myeloid leukocytes fostering occlusive DVT formation. Transfer of platelets into Hmgb1-/- chimeras showed that this cell type is the major source of HMGB1, exposing reduced HMGB1 on their surface upon activation thereby enhancing the recruitment of monocytes. Activated leukocytes in turn support oxidation of HMGB1 unleashing its prothrombotic activity and promoting platelet aggregation. This potentiates the amount of HMGB1 and further nurtures the accumulation and activation of monocytes through receptor for advanced glycation end products (RAGE) and Toll-like receptor 2, leading to local delivery of monocyte-derived tissue factor and cytokines. Moreover, disulfide HMGB1 facilitates formation of prothrombotic neutrophil extracellular traps (NETs) mediated by RAGE, exposing additional HMGB1 on their extracellular DNA strands. Eventually, a vicious circle of coagulation and inflammation is set in motion leading to obstructive DVT formation. Therefore, platelet-derived disulfide HMGB1 is a central mediator of the sterile inflammatory process in venous thrombosis and could be an attractive target for an anti-inflammatory approach for DVT prophylaxis.


Subject(s)
Blood Platelets/metabolism , HMGB1 Protein/physiology , Venous Thrombosis/genetics , Animals , Blood Platelets/pathology , Disulfides/chemistry , Disulfides/metabolism , HMGB1 Protein/chemistry , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Receptor for Advanced Glycation End Products/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Venous Thrombosis/metabolism , Venous Thrombosis/pathology
8.
Bioconjug Chem ; 28(2): 524-538, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28068077

ABSTRACT

Poly(propyleneimine) glycodendrimers fully modified with maltose units were administered to different cancer cell lines and their effect on cell viability was evaluated by using MTS assay and flow cytometry. The mechanism of dendrimer-cell interactions was investigated by the electron paramagnetic resonance (EPR) technique by using a new nitroxide-conjugated glycodendrimer. The nitroxide groups did not modify both the biological properties (cell viability and apoptosis degree) of the dendrimers in the presence of the cells and the dendrimer-cell interactions. Since this class of dendrimers is already known to be biocompatible for human healthy cells, noncancer cells such as human peripheral blood mononuclear cells (PBMCs) and macrophages were also treated with the glycodendrimer, and EPR spectra of the nitroxide-conjugated glycodendrimer were compared for cancer and noncancer cells. It was found that this dendrimer selectively affects the cell viability of tumor cells, while, surprisingly, PBMC proliferation is induced. Moreover, H-bond-active glycodendrimer-cell interactions were different for the different cancer cell lines and noncancer cells. The nitroxide-conjugated glycodendrimer was able to interact with the cell membrane and eventually cross it, getting in contact with cytosol antioxidants. This study helps to clarify the potential anticancer effect of this class of dendrimers opening to future applications of these macromolecules as new antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Dendrimers/pharmacology , Nitrogen Oxides/pharmacology , Polypropylenes/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dendrimers/chemistry , Electron Spin Resonance Spectroscopy , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Maltose/analogs & derivatives , Maltose/pharmacology , Neoplasms/drug therapy , Nitrogen Oxides/chemistry , Polypropylenes/chemistry
9.
Blood ; 125(14): 2254-64, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25575540

ABSTRACT

We present a programmable bioengineered 3-dimensional silk-based bone marrow niche tissue system that successfully mimics the physiology of human bone marrow environment allowing us to manufacture functional human platelets ex vivo. Using stem/progenitor cells, megakaryocyte function and platelet generation were recorded in response to variations in extracellular matrix components, surface topography, stiffness, coculture with endothelial cells, and shear forces. Millions of human platelets were produced and showed to be functional based on multiple activation tests. Using adult hematopoietic progenitor cells our system demonstrated the ability to reproduce key steps of thrombopoiesis, including alterations observed in diseased states. A critical feature of the system is the use of natural silk protein biomaterial allowing us to leverage its biocompatibility, nonthrombogenic features, programmable mechanical properties, and surface binding of cytokines, extracellular matrix components, and endothelial-derived proteins. This in turn offers new opportunities for the study of blood component production ex vivo and provides a superior tissue system for the study of pathologic mechanisms of human platelet production.


Subject(s)
Blood Platelets/cytology , Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Primary Myelofibrosis/pathology , Silk/chemistry , Tissue Scaffolds/chemistry , Adult , Animals , Blood Platelets/metabolism , Bombyx , Bone Marrow Cells/metabolism , Cells, Cultured , Coculture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Matrix , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/metabolism , Primary Myelofibrosis/metabolism , Thrombopoiesis/physiology , Tissue Engineering
10.
Biochim Biophys Acta Gen Subj ; 1861(2): 354-364, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27913191

ABSTRACT

BACKGROUND: The membrane-bound isoform of the receptor for advanced glycation end products (FL-RAGE) is primarily expressed by alveolar epithelial cells and undergoes shedding by the protease ADAM10, giving rise to soluble cleaved RAGE (cRAGE). RAGE has been associated with the pathogenesis of several acute and chronic lung disorders. Whether the proteolysis of FL-RAGE is altered by a given inflammatory stimulus is unknown. Pseudomonas aeruginosa causes nosocomial infections in hospitalized patients and is the major pathogen associated with chronic lung diseases. METHODS: P. aeruginosa was injected in Rage-/- and wild-type mice and the impact on RAGE expression and shedding, levels of inflammation and bacterial growth was determined. RESULTS: Acute P. aeruginosa lung infection in mice induces a reduction of the active form of ADAM10, which determines an increase of FL-RAGE expression on alveolar cells and a concomitant decrease of pulmonary cRAGE levels. This was associated with massive recruitment of leukocytes and release of pro-inflammatory factors, tissue damage and relocation of cRAGE in the alveolar and bronchial cavities. The administration of sRAGE worsened bacterial burden and neutrophils infiltration. RAGE genetic deficiency reduced the susceptibility to P. aeruginosa infection, mitigating leukocyte recruitment, inflammatory molecules production, and bacterial growth. CONCLUSIONS: These data are the first to suggest that inhibition of FL-RAGE shedding, by affecting the FL-RAGE/cRAGE levels, is a novel mechanism for controlling inflammation to acute P. aeruginosa pneumonia. sRAGE in the alveolar space sustains inflammation in this setting. GENERAL SIGNIFICANCE: RAGE shedding may determine the progression of inflammatory lung diseases.


Subject(s)
Inflammation/metabolism , Lung/metabolism , Lung/microbiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Receptor for Advanced Glycation End Products/metabolism , ADAM10 Protein/metabolism , Animals , Glycation End Products, Advanced/metabolism , Inflammation/microbiology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism
11.
Biomolecules ; 14(7)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062550

ABSTRACT

Polyphenols are natural bioactives occurring in medicinal and aromatic plants and food and beverages of plant origin. Compared with conventional therapies, plant-derived phytochemicals are more affordable and accessible and have no toxic side effects. Thus, pharmaceutical research is increasingly inclined to discover and study new and innovative natural molecules for the treatment of several chronic human diseases, like type 2 diabetes mellitus (T2DM) and osteoporosis. These pathological conditions are characterized by a chronic inflammatory state and persistent oxidative stress, which are interconnected and lead to the development and worsening of these two health disorders. Oral nano delivery strategies have been used to improve the bioavailability of polyphenols and to allow these natural molecules to exert their antioxidant, anti-inflammatory, anti-diabetic, and pro-osteogenic biological activities in in vivo experimental models and in patients. Polyphenols are commonly used in the formulations of nutraceuticals, which can counteract the detrimental effects of T2DM and osteoporosis pathologies. This review describes the polyphenols that can exert protective effects against T2DM and osteoporosis through the modulation of specific molecular markers and pathways. These bioactives could be used as adjuvants, in combination with synthetic drugs, in the future to develop innovative therapeutic strategies for the treatment of T2DM and osteoporosis.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Diabetes Mellitus, Type 2 , Hypoglycemic Agents , Osteoporosis , Polyphenols , Humans , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Polyphenols/pharmacology , Polyphenols/chemistry , Polyphenols/therapeutic use , Osteoporosis/drug therapy , Osteoporosis/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Antioxidants/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Animals , Osteogenesis/drug effects , Oxidative Stress/drug effects , Chronic Disease
12.
Neurobiol Dis ; 58: 102-14, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23523633

ABSTRACT

Toll-like receptor 4 (TLR4) activation in neuron and astrocytes by High Mobility Group Box 1 (HMGB1) protein is a key mechanism of seizure generation. HMGB1 also activates the Receptor for Advanced Glycation Endproducts (RAGE), but it was unknown whether RAGE activation contributes to seizures or to HMGB1 proictogenic effects. We found that acute EEG seizures induced by 7ng intrahippocampal kainic acid (KA) were significantly reduced in Rage-/- mice relative to wild type (Wt) mice. The proictogenic effect of HMGB1 was decreased in Rage-/- mice, but less so, than in Tlr4-/- mice. In a mouse mesial temporal lobe epilepsy (mTLE) model, status epilepticus induced by 200ng intrahippocampal KA and the onset of the spontaneous epileptic activity were similar in Rage-/-, Tlr4-/- and Wt mice. However, the number of hippocampal paroxysmal episodes and their duration were both decreased in epileptic Rage-/- and Tlr4-/- mice vs Wt mice. All strains of epileptic mice displayed similar cognitive deficits in the novel object recognition test vs the corresponding control mice. CA1 neuronal cell loss was increased in epileptic Rage-/- vs epileptic Wt mice, while granule cell dispersion and doublecortin (DCX)-positive neurons were similarly affected. Notably, DCX neurons were preserved in epileptic Tlr4-/- mice. We did not find compensatory changes in HMGB1-related inflammatory signaling nor in glutamate receptor subunits in Rage-/- and Tlr4-/- naïve mice, except for ~20% NR2B subunit reduction in Rage-/- mice. RAGE was induced in neurons, astrocytes and microvessels in human and experimental mTLE hippocampi. We conclude that RAGE contributes to hyperexcitability underlying acute and chronic seizures, as well as to the proictogenic effects of HMGB1. RAGE and TLR4 play different roles in the neuropathologic sequelae developing after status epilepticus. These findings reveal new molecular mechanisms underlying seizures, cell loss and neurogenesis which involve inflammatory pathways upregulated in human epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Gene Expression Regulation/genetics , Receptors, Immunologic/metabolism , Seizures/metabolism , Up-Regulation/physiology , Animals , Cell Death/drug effects , Cell Death/genetics , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Electric Stimulation/adverse effects , Electroencephalography , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/pathology , Excitatory Amino Acid Agonists/toxicity , Gene Expression Regulation/drug effects , HMGB1 Protein/administration & dosage , HMGB1 Protein/metabolism , Hippocampus/drug effects , Hippocampus/physiology , Humans , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Seizures/chemically induced , Seizures/etiology , Toll-Like Receptor 4/deficiency , Up-Regulation/genetics
13.
Biofactors ; 49(6): 1205-1222, 2023.
Article in English | MEDLINE | ID: mdl-37409789

ABSTRACT

Reduction in oxygen levels is a key feature in the physiology of the bone marrow (BM) niche where hematopoiesis occurs. The BM niche is a highly vascularized tissue and endothelial cells (ECs) support and regulate blood cell formation from hematopoietic stem cells (HSCs). While in vivo studies are limited, ECs when cultured in vitro at low O2 (<5%), fail to support functional HSC maintenance due to oxidative environment. Therefore, changes in EC redox status induced by antioxidant molecules may lead to alterations in the cellular response to hypoxia likely favoring HSC self-renewal. To evaluate the impact of redox regulation, HUVEC, exposed for 1, 6, and 24 h to 3% O2 were treated with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152). Metabolomic analyses revealed that I-152 increased glutathione levels and influenced the metabolic profiles interconnected with the glutathione system and the redox couples NAD(P)+/NAD(P)H. mRNA analysis showed a lowered gene expression of HIF-1α and VEGF following I-152 treatment whereas TRX1 and 2 were stimulated. Accordingly, the proteomic study revealed the redox-dependent upregulation of thioredoxin and peroxiredoxins that, together with the glutathione system, are the main regulators of intracellular ROS. Indeed, a time-dependent ROS production under hypoxia and a quenching effect of the molecule were evidenced. At the secretome level, the molecule downregulated IL-6, MCP-1, and PDGF-bb. These results suggest that redox modulation by I-152 reduces oxidative stress and ROS level in hypoxic ECs and may be a strategy to fine-tune the environment of an in vitro BM niche able to support functional HSC maintenance.


Subject(s)
Endothelial Cells , NAD , Humans , Reactive Oxygen Species/metabolism , Endothelial Cells/metabolism , NAD/metabolism , Proteomics , Oxidation-Reduction , Hypoxia , Thioredoxins/genetics , Thioredoxins/metabolism , Glutathione/metabolism , Oxygen/metabolism , Sulfur Compounds , Sulfhydryl Compounds
14.
Biomater Sci ; 11(9): 3252-3268, 2023 May 02.
Article in English | MEDLINE | ID: mdl-36939172

ABSTRACT

The application of superparamagnetic iron oxide nanoparticles (SPIONs) in drug delivery, magnetic resonance imaging, cell tracking, and hyperthermia has been long exploited regarding their inducible magnetic properties. Nevertheless, SPIONs remain rapidly cleared from the circulation by the reticuloendothelial system (RES) or mononuclear phagocyte system, with uptake dependent on several factors such as the hydrodynamic diameter, electrical charge and surface coating. This rapid clearance of SPION-based theranostic agents from circulation is one of the main challenges hampering the medical applications that differ from RES targeting. This work proposes a strategy to render biocompatible SPIONs through their encapsulation in the red blood cells (RBCs). In this work, the research has been focused on the multi-step optimization of chemical synthesis of magnetic nanoparticles (MNPs), precisely iron oxide nanoparticles (IONPs) and zinc manganese-ferrite nanoparticles (Zn/Mn FNPs), for encapsulation in human and murine RBCs. The encapsulation through the transient opening of RBC membrane pores requires extensive efforts to deliver high-quality nanoparticles in terms of chemical properties, morphology, stability and biocompatibility. After reaching this goal, in vitro experiments were performed with selected nanomaterials to investigate the potential of engineered MNP-RBC constructs in theranostic approaches.


Subject(s)
Magnetite Nanoparticles , Mice , Animals , Humans , Magnetite Nanoparticles/chemistry , Precision Medicine , Magnetic Resonance Imaging/methods , Drug Delivery Systems , Erythrocytes/metabolism , Theranostic Nanomedicine/methods
15.
BMC Pharmacol ; 11: 8, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21791059

ABSTRACT

BACKGROUND: Since its recent discovery, interleukin-23 has been shown to be involved in the pathogenesis of autoimmune diseases favoring the development of a T cell subset referred to as T helper 17. Glucocorticoids are widely employed in inflammatory and autoimmune diseases as they inhibit pro-inflammatory signaling and prevent production of inflammation mediators. Very limited information is available about the efficacy of synthetic glucocorticoids in containing the expression of interleukin-23 under cell activation. RESULTS: We demonstrate here that the glucocorticoid analogue dexamethasone administered to human monocyte-derived macrophages is indeed able to restrain the expression of interleukin-23 once it has been triggered by a pro-inflammatory stimulus. This effect of dexamethasone is here demonstrated being secondary to suppression of p38 MAPK activity, and involving a protein phosphatase--likely MAPK phosphatase-1 (MKP-1). CONCLUSIONS: Results reported in this paper show that a 10 nanomolar dose of dexamethasone not only prevents inflammatory activation but is also efficacious in confining active inflammation. This effect is here demonstrated not to occur through "canonical" inhibition of the NF-κB transcription factor but through a distinct cascade of down-modulation, that underlines the importance of the transactivating activity of glucocorticoid receptor in the context of its anti-inflammatory action.


Subject(s)
Dexamethasone/pharmacology , Gene Expression/drug effects , Interleukin-23 Subunit p19/genetics , Macrophages/drug effects , Adult , Blotting, Western , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Dose-Response Relationship, Drug , Dual Specificity Phosphatase 1/metabolism , Enzyme-Linked Immunosorbent Assay , Glucocorticoids/pharmacology , Humans , Interleukin-23 Subunit p19/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Front Physiol ; 12: 632682, 2021.
Article in English | MEDLINE | ID: mdl-33679443

ABSTRACT

In the bone marrow (BM) hematopoietic niche, the oxygen tension is usually very low. Such condition affects stem and progenitor cell proliferation and differentiation and, at cellular level regulates hematopoietic growth factors, chemokines and adhesion molecules expression. In turn, these molecules affect the proliferation and maturation of other cellular components of the niche. Due to the complexity of the system we started the in vitro investigations of the IL-6, IL-8, TNFα cytokines expression and the vascular endothelial growth factor (VEGF), considered key mediators of the hematopoietic niche, in human macrophages and macrophage cell line. Since in the niche the oxygen availability is mediated by red blood cells (RBCs), we have influenced the anoxic cell cultures by the administration of oxygenated or deoxygenated RBCs (deoxy RBCs). The results reported in this brief paper show that the presence of RBCs up-regulates IL-8 mRNA while IL-6 and VEGF mRNA expression appears down-regulated. This does not occur when deoxy RBCs are used. Moreover, it appears that the administration of RBCs leads to an increase of TNFα expression levels in MonoMac 6 (MM6). Interestingly, the modulation of these factors likely occurs in a hypoxia-inducible factor-1α (HIF-1α) independent manner. Considering the role of oxygen in the hematopoietic niche further studies should explore these preliminary observations in more details.

17.
Mol Cell Biochem ; 345(1-2): 105-18, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20721684

ABSTRACT

The redox state of the cysteine-rich region of the HIV Tat protein is known to play a crucial role in Tat biological activity. In this article, we show that Tat displays two alternative functional states depending on the presence of either one or three reduced sulphydryl groups in the cysteine-rich region, respectively. Using different approaches, a disulfide pattern has been defined for the Tat protein and a specific DTT-dependent breaking order of disulfide bonds highlighted. The Tat redox state deeply influences macrophage protein uptake. Immunoistochemistry analysis shows that the oxidized protein does not enter cells, whereas partially reduced protein reaches the cytosol and, to a limited extent, the nucleus. Finally electrophoretic analysis shows Tat high-molecular weight multi-aggregation, resulting in the loss of biological activity. This is due to strong electrostatic and metal-binding interactions, whereas Tat dimerization involves metal-binding interactions as well as disulfide bond formation.


Subject(s)
Gene Products, tat/chemistry , Gene Products, tat/pharmacokinetics , Macrophages/metabolism , Protein Multimerization , Cells, Cultured , Chromatography, High Pressure Liquid , Cysteine/metabolism , Disulfides , Endocytosis , Gene Products, tat/metabolism , Humans , Mass Spectrometry , Models, Molecular , Oxidation-Reduction , Protein Transport
18.
Turk Pediatri Ars ; 55(Suppl 1): 85-97, 2020.
Article in English | MEDLINE | ID: mdl-32963484

ABSTRACT

It is described as the eight small European Countries Initiative. The initiative developed during the 63rd session of the World Health Organization Regional Committee for Europe, held in 2013 in Istanbul, Turkey. Eight European countries counting a population of less than 1 million, gathered together under the auspices of the World Health Organization, to form the European Small Countries Initiative for Health. The eight countries include Andorra, Cyprus, Iceland, Luxembourg, Malta, Monaco, Montenegro, and San Marino. The main aim of the small countries network is to foster a common political commitment, useful to develop locally good health practices. A specific goal was the implementation of the Health 2020 European policy framework and strategy for the 21st century, in the context of countries with small populations. The rational is in fact, that countries with smaller populations have a significant advantage to promote and implement policies and strategies for health and well-being that draw on the contribution of many sectors. The eight small European Countries Initiative particularly aims at amplifying the voice of small countries in European and global health contexts, reaching out to local and international legislators and rulers. It further aims at sharing existing resources among members, with the intent to maximize assets, and innovating and applying solutions to increase capacity to improve health. The founding principle of the eight countries initiative network, is that the experiences of small countries can provide useful learning opportunities, particularly in the healthcare area, that can then be used at regional level in more populous nations.

19.
Nutrients ; 12(9)2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32825564

ABSTRACT

This review summarizes the latest advancements in phytochemicals as functional antiviral agents. We focused on flavonoids, like apigenin, vitexin, quercetin, rutin and naringenin, which have shown a wide range of biological effects including antiviral activities. The molecular mechanisms of their antiviral effects mainly consist in the inhibition of viral neuraminidase, proteases and DNA/RNA polymerases, as well as in the modification of various viral proteins. Mixtures of different flavonoids or combination of flavonoids with antiviral synthetic drugs provide an enhancement of their antiviral effects. Recent strategies in drug delivery significantly contribute to overcoming the low bioavailability of flavonoids. Frequent viral infections worldwide have led to the need for new effective antiviral agents, which can be identified among the various phytochemicals. In this light, screening the antiviral activities of a cocktail of flavonoids would be advantageous in order to prevent viral infections and improve current antiviral therapies.


Subject(s)
Antiviral Agents , Drug Delivery Systems , Flavonoids/administration & dosage , Flavonoids/pharmacology , Apigenin/chemistry , Apigenin/pharmacology , Biological Availability , Drug Combinations , Drug Evaluation, Preclinical , Drug Synergism , Flavanones/chemistry , Flavanones/pharmacology , Flavonoids/chemistry , Flavonoids/pharmacokinetics , Humans , Neuraminidase/antagonists & inhibitors , Quercetin/chemistry , Quercetin/pharmacology , Rutin/chemistry , Rutin/pharmacology , Viral Protease Inhibitors , Viral Proteins/metabolism , Virus Diseases/drug therapy , Virus Diseases/prevention & control , Virus Diseases/virology , Viruses/enzymology , Viruses/metabolism
20.
Nanomedicine (Lond) ; 15(8): 739-753, 2020 04.
Article in English | MEDLINE | ID: mdl-32207374

ABSTRACT

Aim: Magnetic particle imaging (MPI) is highly promising for biomedical applications, but optimal tracers for MPI, namely superparamagnetic iron oxide-based contrast agents, are still lacking. Materials & methods: The encapsulation of commercially available nanoparticles, specifically synomag®-D and perimag®, into human red blood cells (RBCs) was performed by a hypotonic dialysis and isotonic resealing procedure. The amounts of superparamagnetic iron oxide incorporated into RBCs were determined by Fe quantification using nuclear magnetic resonance and magnetic particle spectroscopy. Results: Perimag-COOH nanoparticles were identified as the best nanomaterial for encapsulation in RBCs. Perimag-COOH-loaded RBCs proved to be viable cells showing a good magnetic particle spectroscopy performance, while the magnetic signal of synomag-D-COOH-loaded RBCs dropped sharply. Conclusion: Perimag-COOH-loaded RBCs could be a potential tool for MPI diagnostic applications.


Subject(s)
Contrast Media , Erythrocytes , Magnetite Nanoparticles , Humans , Magnetic Resonance Imaging , Magnetics
SELECTION OF CITATIONS
SEARCH DETAIL