Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Int J Mol Sci ; 25(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38339053

ABSTRACT

The blood-retinal barrier (BRB) is strongly compromised in diabetic retinopathy (DR) due to the detachment of pericytes (PCs) from retinal microvessels, resulting in increased permeability and impairment of the BRB. Western blots, immunofluorescence and ELISA were performed on adipose mesenchymal stem cells (ASCs) and pericyte-like (P)-ASCs by co-cultured human retinal endothelial cells (HRECs) under hyperglycemic conditions (HG), as a model of DR. Our results demonstrated that: (a) platelet-derived growth factor receptor (PDGFR) and its activated form were more highly expressed in monocultured P-ASCs than in ASCs, and this expression increased when co-cultured with HRECs under high glucose conditions (HG); (b) the transcription factor Nrf2 was more expressed in the cytoplasmic fraction of ASCs and in the P-ASC nuclear fraction, under normal glucose and, even more, under HG conditions; (c) cytosolic phospholipase A2 activity and prostaglandin E2 release, stimulated by HG, were significantly reduced in P-ASCs co-cultured with HRECs; (d) HO-1 protein content was significantly higher in HG-P-ASCs/HRECs than P-ASCs/HRECs; and (e) VEGF-A levels in media from HG-co-cultures were reduced in P-ASCs/HRECs with respect to ASCs/HRECs. The data obtained highlighted the potential of autologous differentiated ASCs in future clinical applications based on cell therapy to counteract the damage induced by DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Mesenchymal Stem Cells , Humans , Diabetic Retinopathy/therapy , Diabetic Retinopathy/metabolism , Pericytes/metabolism , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Retina/metabolism , Mesenchymal Stem Cells/metabolism , Glucose/metabolism , Cells, Cultured , Diabetes Mellitus/metabolism
2.
Int J Mol Sci ; 24(2)2023 Jan 04.
Article in English | MEDLINE | ID: mdl-36674425

ABSTRACT

Diabetic retinopathy (DR) is characterized by morphologic and metabolic alterations in endothelial cells (ECs) and pericytes (PCs) of the blood-retinal barrier (BRB). The loss of interendothelial junctions, increased vascular permeability, microaneurysms, and finally, EC detachment are the main features of DR. In this scenario, a pivotal role is played by the extensive loss of PCs. Based on previous results, the aim of this study was to assess possible beneficial effects exerted by adipose mesenchymal stem cells (ASCs) and their pericyte-like differentiated phenotype (P-ASCs) on human retinal endothelial cells (HRECs) in high glucose conditions (25 mM glucose, HG). P-ASCs were more able to preserve BRB integrity than ASCs in terms of (a) increased transendothelial electrical resistance (TEER); (b) increased expression of adherens junction and tight junction proteins (VE-cadherin and ZO-1); (c) reduction in mRNA levels of inflammatory cytokines TNF-α, IL-1ß, and MMP-9; (d) reduction in the angiogenic factor VEGF and in fibrotic TGF-ß1. Moreover, P-ASCs counteracted the HG-induced activation of the pro-inflammatory phospho-ERK1/2/phospho-cPLA2/COX-2 pathway. Finally, crosstalk between HRECs and ASCs or P-ASCs based on the PDGF-B/PDGFR-ß axis at the mRNA level is described herein. Thus, P-ASCs might be considered valuable candidates for therapeutic approaches aimed at countering BRB disruption in DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Mesenchymal Stem Cells , Humans , Diabetic Retinopathy/metabolism , Pericytes/metabolism , Endothelial Cells/metabolism , Retina/metabolism , Mesenchymal Stem Cells/metabolism , Blood-Retinal Barrier/metabolism , Glucose/metabolism , RNA, Messenger/metabolism , Diabetes Mellitus/metabolism
3.
Int J Mol Sci ; 23(24)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36555432

ABSTRACT

The stria vascularis (SV) contributes to cochlear homeostasis and consists of three layers, one of which contains the blood-labyrinthic barrier (BLB), with a large number of bovine cochlear pericytes (BCPs). Cisplatin is a chemotherapeutic drug that can damage the SV and cause hearing loss. In this study, cell viability, proliferation rate, cytotoxicity and reactive oxygen species production were evaluated. The protein content of phospho-extracellular signal-regulated kinases (ERK) 1/2, total ERK 1/2, phospho-cytosolic phospholipase A2 (cPLA2), total cPLA2 and cyclooxygenase 2 (COX-2) and the release of prostaglandin E2 (PGE2) and vascular endothelial growth factor (VEGF) from BCPs were analyzed. Finally, the protective effect of platelet-derived growth factor (PDGF-BB) on BCPs treated with cisplatin was investigated. Cisplatin reduced viability and proliferation, activated ERK 1/2, cPLA2 and COX-2 expression and increased PGE2 and VEGF release; these effects were reversed by Dexamethasone. The presence of PDGF-BB during the treatment with cisplatin significantly increased the proliferation rate. No studies on cell regeneration in ear tissue evaluated the effect of the PDGF/Dex combination. The aim of this study was to investigate the effects of cisplatin on cochlear pericytes and propose new otoprotective agents aimed at preventing the reduction of their vitality and thus maintaining the BLB structure.


Subject(s)
Pericytes , Stria Vascularis , Animals , Cattle , Stria Vascularis/metabolism , Cisplatin/toxicity , Cisplatin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Becaplermin/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Platelet-Derived Growth Factor/metabolism
4.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33925714

ABSTRACT

A pericyte-like differentiation of human adipose-derived mesenchymal stem cells (ASCs) was tested in in vitro experiments for possible therapeutic applications in cases of diabetic retinopathy (DR) to replace irreversibly lost pericytes. For this purpose, pericyte-like ASCs were obtained after their growth in a specific pericyte medium. They were then cultured in high glucose conditions to mimic the altered microenvironment of a diabetic eye. Several parameters were monitored, especially those particularly affected by disease progression: cell proliferation, viability and migration ability; reactive oxygen species (ROS) production; inflammation-related cytokines and angiogenic factors. Overall, encouraging results were obtained. In fact, even after glucose addition, ASCs pre-cultured in the pericyte medium (pmASCs) showed high proliferation rate, viability and migration ability. A considerable increase in mRNA expression levels of the anti-inflammatory cytokines transforming growth factor-ß1 (TGF-ß1) and interleukin-10 (IL-10) was observed, associated with reduction in ROS production, and mRNA expression of pro-inflammatory cytokines interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α), and angiogenic factors. Finally, a pmASC-induced better organization of tube-like formation by retinal endothelial cells was observed in three-dimensional co-culture. The pericyte-like ASCs obtained in these experiments represent a valuable tool for the treatment of retinal damages occurring in diabetic patients.


Subject(s)
Glucose/metabolism , Mesenchymal Stem Cells/drug effects , Pericytes/metabolism , Adipose Tissue/metabolism , Adult , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Diabetic Retinopathy/metabolism , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Italy , Mesenchymal Stem Cells/metabolism , Retina/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
5.
Int J Mol Sci ; 21(15)2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32756477

ABSTRACT

Vascular pericytes are an important cellular component in the tumor microenvironment, however, their role in supporting cancer invasion is poorly understood. We hypothesized that PDGF-BB could be involved in the transition of human retinal pericytes (HRPC) in cancer-activated fibroblasts (CAF), induced by the 92.1 uveal melanoma (UM) cell line. In our model system, HRPC were conditioned by co-culturing with 92.1UM for 6 days (cHRPC), in the presence or absence of imatinib, to block PDGF receptor-ß (PDGFRß). The effects of the treatments were tested by wound healing assay, proliferation assay, RT-PCR, high-content screening, Western blot analysis, and invasion assay. Results showed profound changes in cHRPC shape, with increased proliferation and motility, reduction of NG2 and increase of TGF-ß1, α-SMA, vimentin, and FSP-1 protein levels, modulation of PDGF isoform mRNA levels, phospho-PDGFRß, and PDGFRß, as well as phospho-STAT3 increases. A reduction of IL-1ß and IFNγ and an increase in TNFα, IL10, and TGF-ß1, CXCL11, CCL18, and VEGF mRNA in cHRPC were found. Imatinib was effective in preventing all the 92.1UM-induced changes. Moreover, cHRPC elicited a significant increase of 92.1UM cell invasion and active MMP9 protein levels. Our data suggest that retinal microvascular pericytes could promote 92.1UM growth through the acquisition of the CAF phenotype.


Subject(s)
Becaplermin/genetics , Melanoma/metabolism , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Uveal Neoplasms/metabolism , Cancer-Associated Fibroblasts/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate/pharmacology , Matrix Metalloproteinase 9/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Neoplasm Proteins/genetics , Pericytes/drug effects , Pericytes/pathology , Retina/metabolism , Retina/pathology , Transforming Growth Factor beta1/genetics , Tumor Microenvironment/drug effects , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Wound Healing
6.
Int J Mol Sci ; 21(20)2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33065984

ABSTRACT

Early blood retinal barrier (BRB) dysfunction induced by hyperglycemia was related to increased pro-inflammatory activity of phospholipase A2 (PLA2) and the upregulation of vascular endothelial growth factor A (VEGF-A). Here, we tested the role of VEGF-A in high glucose (HG)-induced damage of human retinal endothelial cells (HRECs) mediated by Ca++-dependent (cPLA2) and Ca++-independent (iPLA2) PLA2s. HRECs were treated with normal glucose (5 mM, NG) or high glucose (25 mM, HG) for 48 h with or without the VEGF-trap Aflibercept (Afl, 40 µg/mL), the cPLA2 inhibitor arachidonoyl trifluoromethyl ketone (AACOCF3; 15 µM), the iPLA2 inhibitor bromoenol lactone (BEL; 5 µM), or VEGF-A (80 ng/mL). Both Afl and AACOCF3 prevented HG-induced damage (MTT and LDH release), impairment of angiogenic potential (tube-formation), and expression of VEGF-A mRNA. Furthermore, Afl counteracted HG-induced increase of phospho-ERK and phospho-cPLA2 (immunoblot). VEGF-A in HG-medium increased glucose toxicity, through upregulation of phospho-ERK, phospho-cPLA2, and iPLA2 (about 55%, 45%, and 50%, respectively); immunocytochemistry confirmed the activation of these proteins. cPLA2 knockdown by siRNA entirely prevented cell damage induced by HG or by HG plus VEGF-A, while iPLA2 knockdown produced a milder protective effect. These data indicate that VEGF-A mediates the early glucose-induced damage in retinal endothelium through the involvement of ERK1/2/PLA2 axis activation.


Subject(s)
Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Phospholipases A2/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Arachidonic Acids/pharmacology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Glucose/toxicity , Humans , Phospholipase A2 Inhibitors/pharmacology , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology
7.
J Cell Physiol ; 234(3): 1978-1986, 2019 03.
Article in English | MEDLINE | ID: mdl-30317595

ABSTRACT

The study of strial pericytes has gained great interest as they are pivotal for the physiology of stria vascularis. To provide an easily accessible in vitro model, here we described a growth medium-based approach to obtain and cultivate primary bovine cochlear pericytes (BCP) from the stria vascularis of explanted bovine cochleae. We obtained high-quality pericytes in 8-10 days with a > 90% purity after the second passage. Immunocytochemical analysis showed a homogeneous population of cells expressing typical pericyte markers, such as neural/glial antigen 2 (NG2), platelet-derived growth factor receptorß (PDGFRß), α-smooth muscle actin (α-SMA), and negative for the endothelial marker von Willebrand factor. When challenged with tumor necrosis factor or lipopolysaccharide, BCP changed their shape, similarly to human retinal pericytes (HRPC). The sensitivity of BCP to ototoxic drugs was evaluated by challenging with cisplatin or gentamicin for 48 hr. Compared to human retinal endothelial cells and HRPC, cell viability of BCP was significantly lower ( p < 0.05) after the treatment with gentamicin or cisplatin. These data indicate that our protocol provides a simple and reliable method to obtain highly pure strial BCP. Furthermore, BCP are suitable to assess the safety profile of molecules which supposedly exert ototoxic activity, and may represent a valid alternative to in vivo tests.


Subject(s)
Cochlea/cytology , Pericytes/cytology , Stria Vascularis/cytology , Actins/metabolism , Animals , Antigens/metabolism , Biomarkers/metabolism , Cattle , Cell Culture Techniques/methods , Cell Survival , Cisplatin/toxicity , Cochlea/drug effects , Cochlea/metabolism , Culture Media , Drug Evaluation, Preclinical/methods , Gentamicins/toxicity , In Vitro Techniques , Models, Biological , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Pericytes/drug effects , Pericytes/metabolism , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stria Vascularis/drug effects , Stria Vascularis/metabolism
8.
Pharmacol Res ; 141: 384-391, 2019 03.
Article in English | MEDLINE | ID: mdl-30648615

ABSTRACT

The dopamine D3 receptor (D3R), in the nucleus accumbens (NAc), plays an important role in alcohol reward mechanisms. The major neuronal type within the NAc is the GABAergic medium spiny neuron (MSN), whose activity is regulated by dopaminergic inputs. We previously reported that genetic deletion or pharmacological blockade of D3R increases GABAA α6 subunit in the ventral striatum. Here we tested the hypothesis that D3R-dependent changes in GABAA α6 subunit in the NAc affect voluntary alcohol intake, by influencing the inhibitory transmission of MSNs. We performed in vivo and ex vivo experiments in D3R knockout (D3R -/-) mice and wild type littermates (D3R +/+). Ro 15-4513, a high affinity α6-GABAA ligand was used to study α6 activity. At baseline, NAc α6 expression was negligible in D3R+/+, whereas it was robust in D3R-/-; other relevant GABAA subunits were not changed. In situ hybridization and qPCR confirmed α6 subunit mRNA expression especially in the NAc. In the drinking-in-the-dark paradigm, systemic administration of Ro 15-4513 inhibited alcohol intake in D3R+/+, but increased it in D3R-/-; this was confirmed by intra-NAc administration of Ro 15-4513 and furosemide, a selective α6-GABAA antagonist. Whole-cell patch-clamp showed peak amplitudes of miniature inhibitory postsynaptic currents in NAc medium spiny neurons higher in D3R-/- compared to D3R+/+; Ro 15-4513 reduced the peak amplitude in the NAc of D3R-/-, but not in D3R+/+. We conclude that D3R-dependent enhanced expression of α6 GABAA subunit inhibits voluntary alcohol intake by increasing GABA inhibition in the NAc.


Subject(s)
Binge Drinking/genetics , GABAergic Neurons/pathology , Receptors, Dopamine D3/genetics , Receptors, GABA-A/genetics , Animals , Binge Drinking/pathology , GABAergic Neurons/metabolism , Gene Expression Regulation , Male , Mice , Mice, Knockout , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Protein Subunits/genetics , RNA, Messenger/genetics
9.
Int J Mol Sci ; 20(24)2019 Dec 17.
Article in English | MEDLINE | ID: mdl-31861092

ABSTRACT

Pericytes are branched cells located in the wall of capillary blood vessels that are found throughout the body, embedded within the microvascular basement membrane and wrapping endothelial cells, with which they establish a strong physical contact. Pericytes regulate angiogenesis, vessel stabilization, and contribute to the formation of both the blood-brain and blood-retina barriers by Angiopoietin-1/Tie-2, platelet derived growth factor (PDGF) and transforming growth factor (TGF) signaling pathways, regulating pericyte-endothelial cell communication. Human pericytes that have been cultured for a long period give rise to multilineage progenitor cells and exhibit mesenchymal stem cell (MSC) features. We focused our attention on the roles of pericytes in brain and ocular diseases. In particular, pericyte involvement in brain ischemia, brain tumors, diabetic retinopathy, and uveal melanoma is described. Several molecules, such as adenosine and nitric oxide, are responsible for pericyte shrinkage during ischemia-reperfusion. Anti-inflammatory molecules, such as IL-10, TGFß, and MHC-II, which are increased in glioblastoma-activated pericytes, are responsible for tumor growth. As regards the eye, pericytes play a role not only in ocular vessel stabilization, but also as a stem cell niche that contributes to regenerative processes in diabetic retinopathy. Moreover, pericytes participate in melanoma cell extravasation and the genetic ablation of the PDGF receptor reduces the number of pericytes and aberrant tumor microvessel formation with important implications for therapy efficacy. Thanks to their MSC features, pericytes could be considered excellent candidates to promote nervous tissue repair and for regenerative medicine.


Subject(s)
Brain/physiology , Microvessels/physiology , Pericytes/physiology , Regeneration/physiology , Retina/physiology , Retinal Vessels/physiology , Animals , Blood-Brain Barrier/physiology , Blood-Retinal Barrier/physiology , Brain/blood supply , Humans , Microvessels/cytology , Pericytes/cytology
10.
Pharmacol Res ; 106: 10-20, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26875816

ABSTRACT

The ectopic re-activation of cell cycle in neurons is an early event in the pathogenesis of Alzheimer's disease (AD), which could lead to synaptic failure and ensuing cognitive deficits before frank neuronal death. Cytostatic drugs that act as cyclin-dependent kinase (CDK) inhibitors have been poorly investigated in animal models of AD. In the present study, we examined the effects of flavopiridol, an inhibitor of CDKs currently used as antineoplastic drug, against cell cycle reactivation and memory loss induced by intracerebroventricular injection of Aß1-42 oligomers in CD1 mice. Cycling neurons, scored as NeuN-positive cells expressing cyclin A, were found both in the frontal cortex and in the hippocampus of Aß-injected mice, paralleling memory deficits. Starting from three days after Aß injection, flavopiridol (0.5, 1 and 3mg/kg) was intraperitoneally injected daily, for eleven days. Here we show that a treatment with flavopiridol (0.5 and 1mg/kg) was able to rescue the loss of memory induced by Aß1-42, and to prevent the occurrence of ectopic cell-cycle events in the mouse frontal cortex and hippocampus. This is the first evidence that a cytostatic drug can prevent cognitive deficits in a non-transgenic animal model of AD.


Subject(s)
Amyloid beta-Peptides/adverse effects , Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Memory/drug effects , Peptide Fragments/adverse effects , Piperidines/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory Disorders/etiology , Memory Disorders/metabolism , Mice , Neurons/drug effects , Neurons/metabolism
11.
Nutrients ; 16(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39339713

ABSTRACT

Background/Objectives: Rare diseases are a wide and heterogeneous group of multisystem life-threatening or chronically debilitating clinical conditions with reduced life expectancy and a relevant mortality rate in childhood. Some of these disorders have typical neurological symptoms, presenting from birth to adulthood. Dietary patterns and nutritional compounds play key roles in the onset and progression of neurological disorders, and the impact of alimentary needs must be enlightened especially in rare neurological diseases. This work aims to collect the in vitro, in vivo, and clinical evidence on the effects of diet and of nutrient intake on some rare neurological disorders, including some genetic diseases, and rare brain tumors. Herein, those aspects are critically linked to the genetic, biological, biochemical, and pathophysiological hallmarks typical of each disorder. Methods: By searching the major web-based databases (PubMed, Web of Science Core Collection, DynaMed, and Clinicaltrials.gov), we try to sum up and improve our understanding of the emerging role of nutrition as both first-line therapy and risk factors in rare neurological diseases. Results: In line with the increasing number of consensus opinions suggesting that nutrients should receive the same attention as pharmacological treatments, the results of this work pointed out that a standard dietary recommendation in a specific rare disease is often limited by the heterogeneity of occurrent genetic mutations and by the variability of pathophysiological manifestation. Conclusions: In conclusion, we hope that the knowledge gaps identified here may inspire further research for a better evaluation of molecular mechanisms and long-term effects.


Subject(s)
Diet , Nervous System Diseases , Nutrients , Rare Diseases , Humans , Nervous System Diseases/diet therapy
12.
Front Biosci (Landmark Ed) ; 27(11): 308, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36472115

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) accumulation plays a pivotal role in the onset of cell damage induced by hyperglycemia and represents one of the major factors in the pathogenesis of diabetic retinopathy. In this study, we tested the antioxidants cyanidin-3-O-glucoside (C3G) and verbascoside (Verb) in the protection of retinal endothelium against glucose toxicity "in vitro". METHODS: Increasing amounts (5-50 µM) of C3G, Verb or the combination of both compounds were tested in Human Retinal Endothelial Cells (HREC) grown with normal glucose (5 mM, NG) or high glucose (25 mM, HG). RESULTS: Reduced cell viability and enhanced ROS levels (evaluated by MTT and H2DCFDA assays, respectively) in HG-stimulated HREC were restored by C3G and Verb in a dose-dependent manner, achieving the maximum protection in the presence of both compounds. Moreover, co-treatment with C3G and Verb worked better than each single molecule alone in the prevention of the disruption of blood-retinal-barrier-like properties by HG in a confluent HREC monolayer, as assessed by trans endothelial electrical resistance (TEER) and Na-Fluorescein permeability assays. Accordingly, C3G and Verb together also better counteracted the HG-induced down-regulation of the tight junction membrane proteins Zonula Occludens-1 and VE-Cadherin evaluated by immunocytochemical and Western blot analyses. CONCLUSIONS: In conclusion, our data indicate that C3G and Verb could efficiently protect the retinal endothelium against high glucose damage.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Antioxidants/pharmacology , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Reactive Oxygen Species/metabolism , Endothelial Cells/metabolism , Glucosides/pharmacology , Glucose/pharmacology
13.
Front Pharmacol ; 13: 971164, 2022.
Article in English | MEDLINE | ID: mdl-36091806

ABSTRACT

The impairment of the blood retinal barrier (BRB) represents one of the main features of diabetic retinopathy, a secondary microvascular complication of diabetes. Hyperglycemia is a triggering factor of vascular cells damage in diabetic retinopathy. The aim of this study was to assess the effects of vitamin D3 on BRB protection, and to investigate its regulatory role on inflammatory pathways. We challenged human retinal endothelial cells with high glucose (HG) levels. We found that vitamin D3 attenuates cell damage elicited by HG, maintaining cell viability and reducing the expression of inflammatory cytokines such as IL-1ß and ICAM-1. Furthermore, we showed that vitamin D3 preserved the BRB integrity as demonstrated by trans-endothelial electrical resistance, permeability assay, and cell junction morphology and quantification (ZO-1 and VE-cadherin). In conclusion this in vitro study provided new insights on the retinal protective role of vitamin D3, particularly as regard as the early phase of diabetic retinopathy, characterized by BRB breakdown and inflammation.

14.
Ann Anat ; 244: 151985, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35914630

ABSTRACT

BACKGROUND: Taste buds, the morphofunctional units for taste perception, transduce gustatory stimuli using G protein-coupled receptors and a complex arrangement of ion channels, among which TRPV4, a member of the TRP superfamily. Studies on taste buds development on gilthead seabream are unknown, and the TRPV4 expression on fish taste cells studies were conducted only on zebrafish. METHODS: In our study, we have investigated the histological features of the gilthead seabream tongue dorsal surface from the earliest stage of development using Masson trichrome with aniline blue staining. Additionally, the TRPV4 expression pattern was studied by means of immunohistochemical labeling and quantitative RT-PCR. RESULTS: We have recorded for the first time on gilthead seabream lingual dorsal surface the presence of, stage-specific, three types of taste buds: type I, type II and type III in larvae, juveniles and adults respectively. At 40 days post-hatching, taste buds were mature-looking. TRPV4 expression was detected in a subpopulation of taste cells of larvae, juveniles, and adults. Furthermore, TRPV4 was expressed in the basal epithelial cells of the tongue at the larvae and juvenile stage, while this expression pattern was more diffused within all the epithelial cell layers in the adult. CONCLUSION: Our findings presume a taste sensory role of TRPV4 in the three stage-specific taste buds and oral epithelia of gilthead seabream. In addition to its sensory role on the epithelial cell layers, we hypothesize that TRPV4 is implicated in epithelial cells differentiation and membrane protection.


Subject(s)
Sea Bream , Taste Buds , Animals , Sea Bream/metabolism , TRPV Cation Channels/metabolism , Zebrafish/metabolism , Tongue , Taste Buds/metabolism , Zebrafish Proteins/metabolism
15.
Int J Pharm ; 627: 122195, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36115466

ABSTRACT

Melatonin (MEL) is a pleiotropic neurohormone of increasing interest as a neuroprotective agent in ocular diseases. Improving the mucoadhesiveness is a proposed strategy to increase the bioavailability of topical formulations. Herein, the design and optimization of MEL-loaded lipid-polymer hybrid nanoparticles (mel-LPHNs) using Design of Experiment (DoE) was performed. LPHNs consisted of PLGA-PEG polymer nanoparticles coated with a cationic lipid-shell. The optimized nanomedicine showed suitable size for ophthalmic administration (189.4 nm; PDI 0.260) with a positive surface charge (+39.8 mV), high encapsulation efficiency (79.8 %), suitable pH and osmolarity values, good mucoadhesive properties and a controlled release profile. Differential Scanning Calorimetry and Fourier-Transform Infrared Spectroscopy confirmed the encapsulation of melatonin in the systems and the interaction between lipids and polymer matrix. Biological evaluation in an in vitro model of diabetic retinopathy demonstrated enhanced neuroprotective and antioxidant activities of mel-LPHNs, compared to melatonin aqueous solution at the same concentration (0.1 and 1 µM). A modified Draize test was performed to assess the ocular tolerability of the formulation showing no signs of irritation. To the best our knowledge, this study reported for the first time the development of mel-LPHNs, a novel and safe hybrid platform suitable for the topical management of retinal diseases.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Melatonin , Nanoparticles , Neuroprotective Agents , Humans , Nanomedicine , Melatonin/chemistry , Delayed-Action Preparations , Antioxidants/pharmacology , Diabetic Retinopathy/drug therapy , Nanoparticles/chemistry , Polymers/chemistry , Lipids/chemistry , Particle Size , Drug Carriers/chemistry
16.
Microvasc Res ; 81(1): 1-17, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21094175

ABSTRACT

Glioma is characterized by an active production of proangiogenic molecules. We observed that conditioned medium (CM) from C6 glioma significantly enhanced proliferation and migration of immortalized rat brain GP8.3 endothelial cells (ECs) and primary bovine brain microvascular ECs. The glioma CM effect was significantly reduced by cytosolic (cPLA(2)) and Ca(++)-independent (iPLA(2)) phospholipase A(2), cyclooxygenase-2, and protein kinase inhibitors. In GP8.3 ECs, cPLA(2) and iPLA(2) enzyme activities and phosphorylation of cPLA(2), significantly stimulated after 24h CM co-incubation, were attenuated by PLA(2), PI3-K, MEK-1, and ERK1/2 inhibitors. By confocal microscopy, in glioma CM-stimulated ECs, enhancement of fluorescence signals for phospho-cPLA(2), phospho-ERK1/2, phospho-PKCα, COX-2, and iPLA(2) was in parallel observed. Electroporation of anti-iPLA(2) and cPLA(2) antibodies and siRNAs directed against iPLA(2) and cPLA(2) significantly inhibited cell proliferation and migration. Incubation of CM- or VEGF peptide-stimulated ECs with antibodies against VEGF or VEGFR-1/-2 receptors strongly reduced mitotic rate, cell migration, and phospho-cPLA(2) and iPLA(2) protein levels. The findings suggest that PLA(2) activities are involved in stimulating EC migration and proliferation in the presence of glioma CM and that cPLA(2) is positively regulated upstream by PI3-K, PKCα, and ERK1/2 signal cascades. Our work provides new insights in understanding EC metabolism and signaling during tumor angiogenesis.


Subject(s)
Brain/cytology , Endothelial Cells/enzymology , Glioma/metabolism , Neovascularization, Pathologic/metabolism , Phospholipases A2, Calcium-Independent/metabolism , Phospholipases A2, Cytosolic/metabolism , Animals , Antibodies/administration & dosage , Antibodies/immunology , Antibodies/pharmacology , Cattle , Cell Line, Transformed , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Electroporation , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Gene Expression/genetics , Glioma/blood supply , Intracellular Space/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Phospholipases A2, Calcium-Independent/antagonists & inhibitors , Phospholipases A2, Calcium-Independent/genetics , Phospholipases A2, Calcium-Independent/immunology , Phospholipases A2, Cytosolic/antagonists & inhibitors , Phospholipases A2, Cytosolic/genetics , Phospholipases A2, Cytosolic/immunology , Phosphorylation/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/pharmacology
17.
World J Stem Cells ; 13(6): 632-644, 2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34249232

ABSTRACT

Stem cell-based treatments have been extensively explored in the last few decades to develop therapeutic strategies aimed at providing effective alternatives for those human pathologies in which surgical or pharmacological therapies produce limited effects. Among stem cells of different sources, mesenchymal stem cells (MSCs) offer several advantages, such as the absence of ethical concerns, easy harvesting, low immunogenicity and reduced tumorigenesis risks. Other than a multipotent differentiation ability, MSCs can release extracellular vesicles conveying proteins, mRNA and microRNA. Thanks to these properties, new therapeutic approaches have been designed for the treatment of various pathologies, including ocular diseases. In this review, the use of different MSCs and different administration strategies are described for the treatment of diabetic retinopathy, glaucoma, and retinitis pigmentosa. In a large number of investigations, positive results have been obtained by in vitro experiments and by MSC administration in animal models. Most authors agree that beneficial effects are likely related to MSC paracrine activity. Based on these considerations, many clinical trials have already been carried out. Overall, although some adverse effects have been described, promising outcomes are reported. It can be assumed that in the near future, safer and more effective protocols will be developed for more numerous clinical applications to improve the quality of life of patients affected by eye diseases.

18.
Cells ; 11(1)2021 12 24.
Article in English | MEDLINE | ID: mdl-35011613

ABSTRACT

Hyperglycemia-induced impairment of the blood-retinal barrier represents the main pathological event in diabetic retinopathy that is elicited by a reduced cellular response to an accumulation of reactive oxygen species (ROS) and increased inflammation. The purpose of the study was to evaluate whether the selective ß1-adrenoreceptor (ß1-AR) antagonist metoprolol could modulate the inflammatory response to hyperglycemic conditions. For this purpose, human retinal endothelial cells (HREC) were treated with normal (5 mM) or high glucose (25 mM, HG) in the presence of metoprolol (10 µM), epinephrine (1 µM), or both compounds. Metoprolol prevented both the HG-induced reduction of cell viability (MTT assays) and the modulation of the angiogenic potential of HREC (tube formation assays) reducing the TNF-α, IL-1ß, and VEGF mRNA levels (qRT-PCR). Moreover, metoprolol prevented the increase in phospho-ERK1/2, phospho-cPLA2, COX2, and protein levels (Western blot) as well as counteracting the translocation of ERK1/2 and cPLA2 (high-content screening). Metoprolol reduced ROS accumulation in HG-stimulated HREC by activating the anti-oxidative cellular response mediated by the Keap1/Nrf2/HO-1 pathway. In conclusion, metoprolol exerted a dual effect on HG-stimulated HREC, decreasing the activation of the pro-inflammatory ERK1/2/cPLA2/COX2 axis, and counteracting ROS accumulation by activating the Keap1/Nrf2/HO-1 pathway.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Anti-Inflammatory Agents/pharmacology , Endothelial Cells/pathology , Glucose/toxicity , Metoprolol/pharmacology , Microvessels/pathology , Retina/pathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epinephrine/pharmacology , Heme Oxygenase-1/metabolism , Humans , Interleukin-1beta/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/metabolism , Neovascularization, Physiologic/drug effects , Phospholipases A2, Cytosolic/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
World J Stem Cells ; 13(11): 1783-1796, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34909123

ABSTRACT

BACKGROUND: Adipose-derived stem cells (ASCs) have been increasingly explored for cell-based medicine because of their numerous advantages in terms of easy availability, high proliferation rate, multipotent differentiation ability and low immunogenicity. In this respect, they have been widely investigated in the last two decades to develop therapeutic strategies for a variety of human pathologies including eye disease. In ocular diseases involving the retina, various cell types may be affected, such as Müller cells, astrocytes, photoreceptors and retinal pigment epithelium (RPE), which plays a fundamental role in the homeostasis of retinal tissue, by secreting a variety of growth factors that support retinal cells. AIM: To test ASC neural differentiation using conditioned medium (CM) from an RPE cell line (ARPE-19). METHODS: ASCs were isolated from adipose tissue, harvested from the subcutaneous region of healthy donors undergoing liposuction procedures. Four ASC culture conditions were investigated: ASCs cultured in basal Dulbecco's Modified Eagle Medium (DMEM); ASCs cultured in serum-free DMEM; ASCs cultured in serum-free DMEM/F12; and ASCs cultured in a CM from ARPE-19, a spontaneously arising cell line with a normal karyotype derived from a human RPE. Cell proliferation rate and viability were assessed by crystal violet and MTT assays at 1, 4 and 8 d of culture. At the same time points, ASC neural differentiation was evaluated by immunocytochemistry and western blot analysis for typical neuronal and glial markers: Nestin, neuronal specific enolase (NSE), protein gene product (PGP) 9.5, and glial fibrillary acidic protein (GFAP). RESULTS: Depending on the culture medium, ASC proliferation rate and viability showed some significant differences. Overall, less dense populations were observed in serum-free cultures, except for ASCs cultured in ARPE-19 serum-free CM. Moreover, a different cell morphology was seen in these cultures after 8 d of treatment, with more elongated cells, often showing cytoplasmic ramifications. Immunofluorescence results and western blot analysis were indicative of ASC neural differentiation. In fact, basal levels of neural markers detected under control conditions significantly increased when cells were cultured in ARPE-19 CM. Specifically, neural marker overexpression was more marked at 8 d. The most evident increase was observed for NSE and GFAP, a modest increase was observed for nestin, and less relevant changes were observed for PGP9.5. CONCLUSION: The presence of growth factors produced by ARPE-19 cells in tissue culture induces ASCs to express neural differentiation markers typical of the neuronal and glial cells of the retina.

20.
Cell Death Dis ; 12(10): 905, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34611142

ABSTRACT

Age-related disorders, such as Alzheimer's disease (AD) and age-related macular degeneration (AMD) share common features such as amyloid-ß (Aß) protein accumulation. Retinal deposition of Aß aggregates in AMD patients has suggested a potential link between AMD and AD. In the present study, we analyzed the expression pattern of a focused set of miRNAs, previously found to be involved in both AD and AMD, in the retina of a triple transgenic mouse model of AD (3xTg-AD) at different time-points. Several miRNAs were differentially expressed in the retina of 3xTg-AD mice, compared to the retina of age-matched wild-type (WT) mice. In particular, bioinformatic analysis revealed that miR-155 had a central role in miRNA-gene network stability, regulating several pathways, including apoptotic and inflammatory signaling pathways modulated by TNF-related apoptosis-inducing ligand (TNFSF10). We showed that chronic treatment of 3xTg-AD mice with an anti-TNFSF10 monoclonal antibody was able to inhibit the retinal expression of miR-155, which inversely correlated with the expression of its molecular target SOCS-1. Moreover, the fine-tuned mechanism related to TNFSF10 immunoneutralization was tightly linked to modulation of TNFSF10 itself and its death receptor TNFRSF10B, along with cytokine production by microglia, reactive gliosis, and specific AD-related neuropathological hallmarks (i.e., Aß deposition and Tau phosphorylation) in the retina of 3xTg-AD mice. In conclusion, immunoneutralization of TNFSF10 significantly preserved the retinal tissue in 3xTg-AD mice, suggesting its potential therapeutic application in retinal degenerative disorders.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Inflammation/pathology , MicroRNAs/metabolism , Retina/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Base Sequence , Calcium-Binding Proteins/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Gliosis/pathology , Inflammation/complications , Inflammation/genetics , Interleukin-10/metabolism , Mice, Transgenic , MicroRNAs/genetics , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Phosphorylation/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL