Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Riv Psichiatr ; 58(6): 293-301, 2023.
Article in English | MEDLINE | ID: mdl-38032033

ABSTRACT

INTRODUCTION AND AIMS: Bipolar disorder (BD) is a severe and recurring mental illness associated with a significant personal and social burden. It has been recently hypothesized that increased levels of pro-inflammatory cytokines and cortisol, which is also associated with a reduced expression of the brain-derived neurotrophic factor (BDNF), may influence affective recurrences in BD. Our study aims to: 1) assess changes in the levels of peripheral cytokines, BDNF and salivary cortisol during acute and euthymic phases of bipolar disorder, compared to that of a sample oh healthy controls; 2) evaluate whether these changes represent a biosignature for the different phases of the illness. MATERIALS AND METHODS: Patients aged 18-65 years old, with a diagnosis of BD I or II types, will be enrolled during an acute episode, according to DSM-5 criteria, together with age- and gender-matched healthy controls. Blood and salivary samples will be collected at baseline and after 3 and 6 months. Validated assessment instruments will be administered to all participants for the evaluation of symptom severity, global functioning, suicidal risk, stress levels and physical comorbidities. EXPECTED RESULTS: We expect changes in inflammatory and neuroendocrine indices to be predictive of the onset of an acute phase of bipolar disorder and that overall levels of cytokines, cortisol and BDNF are overall significantly different between BD patients and healthy controls. CONCLUSIONS: The longitudinal design of the study will allow to assess whether the presence of acute affective symptoms in BD patients correlates with significantly higher levels of cytokines and salivary cortisol and with reduced BDNF levels compared to euthymic phases. Moreover, the comparison with healthy control subjects will allow to understand if inflammatory mediators as well as the hypothalamic-pituitary-adrenal (HPA) axis are chronically elevated in BD patients and are independent from mood swings.


Subject(s)
Bipolar Disorder , Adolescent , Adult , Aged , Humans , Middle Aged , Young Adult , Bipolar Disorder/complications , Brain-Derived Neurotrophic Factor , Cytokines , Hydrocortisone , Personality Disorders , Male , Female , Controlled Clinical Trials as Topic
2.
Brain Sci ; 13(8)2023 Aug 20.
Article in English | MEDLINE | ID: mdl-37626577

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays a key role in brain development, contributing to neuronal survival and neuroplasticity. Previous works have found that BDNF is involved in several neurological or psychiatric diseases. In this review, we aimed to collect all available data on BDNF and bipolar disorder (BD) and assess if BDNF could be considered a biomarker for BD. We searched the most relevant medical databases and included studies reporting original data on BDNF circulating levels or Val66Met polymorphism. Only articles including a direct comparison with healthy controls (HC) and patients diagnosed with BD according to international classification systems were included. Of the 2430 identified articles, 29 were included in the present review. Results of the present review show a reduction in BDNF circulating levels during acute phases of BD compared to HC, which increase after effective therapy of the disorders. The Val66Met polymorphism was related to features usually associated with worse outcomes. High heterogeneity has been observed regarding sample size, clinical differences of included patients, and data analysis approaches, reducing comparisons among studies. Although more studies are needed, BDNF seems to be a promising biomarker for BD.

3.
Sci Rep ; 10(1): 12250, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704131

ABSTRACT

Cardiotoxicity remains a serious problem in anthracycline-treated oncologic patients. Therapeutic modulation of microRNA expression is emerging as a cardioprotective approach in several cardiovascular pathologies. MiR-34a increased in animals and patients exposed to anthracyclines and is involved in cardiac repair. In our previous study, we demonstrated beneficial effects of miR-34a silencing in rat cardiac cells exposed to doxorubicin (DOXO). The aim of the present work is to evaluate the potential cardioprotective properties of a specific antimiR-34a (Ant34a) in an experimental model of DOXO-induced cardiotoxicity. Results indicate that in our model systemic administration of Ant34a completely silences miR-34a myocardial expression and importantly attenuates DOXO-induced cardiac dysfunction. Ant34a systemic delivery in DOXO-treated rats triggers an upregulation of prosurvival miR-34a targets Bcl-2 and SIRT1 that mediate a reduction of DOXO-induced cardiac damage represented by myocardial apoptosis, senescence, fibrosis and inflammation. These findings suggest that miR-34a therapeutic inhibition may have clinical relevance to attenuate DOXO-induced toxicity in the heart of oncologic patients.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Doxorubicin/adverse effects , Gene Silencing , Genetic Predisposition to Disease , MicroRNAs/genetics , Animals , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cardiotoxicity/diagnosis , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/genetics , Disease Models, Animal , Doxorubicin/therapeutic use , Fibrosis , Gene Expression Regulation/drug effects , Genes, bcl-2 , Models, Biological , Myocardium/metabolism , Rats , Sirtuin 1/genetics
4.
Biomolecules ; 10(3)2020 02 26.
Article in English | MEDLINE | ID: mdl-32111073

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) are involved in vascular wall degradation, and drugs able to modulate MMP activity can be used to prevent or treat aneurysmal disease. In this study, we evaluated the effects of statins on MMP-2, MMP-9, and neutrophil gelatinase-associated lipocalin (NGAL) in both plasma and tissue in patients with aneurysmal disease. METHODS: We performed a prospective, single-blind, multicenter, control group clinical drug trial on 184 patients of both sexes >18 years old with a diagnosis of arterial aneurysmal disease. Enrolled patients were divided into two groups: Group I under statin treatment and Group II not taking statins. In addition, 122 patients without aneurysmal disease and under statin treatment were enrolled as a control group (Group III). The expression of MMPs and NGAL in plasma was evaluated using ELISA, while their expression in endothelial tissues was evaluated using Western blot. RESULTS: The ELISA test revealed greater plasma levels (p < 0.01) of MMPs and NGAL in Groups I and II vs. Group III. Western blot analysis showed higher expression (p < 0.01) of MMPs and NGAL in Group II vs. Group I, and this increase was significantly higher (p < 0.01) in patients treated with low potency statins compared to high potency ones. CONCLUSIONS: MMPs and NGAL seem to play a major role in the development of aneurysms, and their modulation by statins suggests that these drugs could be used to prevent arterial aneurysmal disease.


Subject(s)
Aneurysm/drug therapy , Aneurysm/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipocalin-2/analysis , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Aged , Aneurysm/blood , Female , Humans , Lipocalin-2/blood , Male , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Middle Aged , Pilot Projects , Prospective Studies
5.
Stem Cells Int ; 2018: 9492038, 2018.
Article in English | MEDLINE | ID: mdl-29731780

ABSTRACT

Pulmonary emphysema is a respiratory condition characterized by alveolar destruction that leads to airflow limitation and reduced lung function. Although with extensive research, the pathophysiology of emphysema is poorly understood and effective treatments are still missing. Evidence suggests that mesenchymal stem cells (MSCs) possess the ability to engraft the injured tissues and induce repair via a paracrine effect. Thus, the aim of this study was to test the effects of the intratracheal administration of lung-derived mouse MSCs in a model of elastase-induced emphysema. Pulmonary function (static lung compliance) showed an increased stiffness induced by elastase, while morphometric findings (mean linear intercept and tissue/alveolar area) confirmed the severity of alveolar disruption. Contrarily, MSC administration partially restored lung elasticity and alveolar architecture. In the absence of evidence that MSCs acquired epithelial phenotype, we detected an increased proliferative activity of aquaporin 5- and surfactant protein C-positive lung cells, suggesting MSC-driven paracrine mechanisms. The data indicate the mediation of hepatocyte growth factor in amplifying MSC-driven tissue response after injury. Our study shed light on supportive properties of lung-derived MSCs, although the full identification of mechanisms orchestrated by MSCs and responsible for epithelial repair after injury is a critical aspect yet to be achieved.

6.
Pharmacol Res ; 127: 4-14, 2018 01.
Article in English | MEDLINE | ID: mdl-28336372

ABSTRACT

Anthracycline cardiotoxicity remains a serious problem in paediatric and adult cancer survivors, and the advancement of cardio-oncology is a necessary step for an effective care of the patients that experience adverse cardiovascular effects. In this review, we discuss the multiple instruments used by clinicians that constitute the current strategies for primary and secondary prevention aiming at contrasting the onset of early and late doxorubicin-induced cardiotoxic events. The importance of early detection of cardiotoxicity and the following pharmacological therapy has been acknowledged with the emphasis put on impaired diastolic function, an increasingly recognized precocious sign of doxorubicin cardiotoxicity with an emerging scientific and clinical interest. We highlight the involvement of progenitor cells of cardiac and extra-cardiac origin as well as multiple cardiac cell types (fibroblasts and vasculature cells), focusing on molecular signals involved in cellular injury and response. Oxidative stress, DNA damage, senescence and cell death are established mechanisms driving anthracycline toxicity, but the comprehension of their relative weight on affecting specific cell type behaviour remains to be consolidated. The contribution of these crucial stressors and the emerging tools for preserving cell function are discussed.


Subject(s)
Cardiotoxicity/prevention & control , Doxorubicin/adverse effects , Biomarkers, Pharmacological , Cardiotoxicity/drug therapy , Humans , Models, Biological
7.
Br J Pharmacol ; 174(21): 3696-3712, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28320043

ABSTRACT

BACKGROUND AND PURPOSE: Doxorubicin is a highly effective anticancer drug, but its clinical application is hampered by cardiotoxicity. Asymptomatic diastolic dysfunction can be the earliest manifestation of doxorubicin cardiotoxicity. Therefore, a search for therapeutic intervention that can interfere with early manifestations and possibly prevent later development of cardiotoxicity is warranted. Increased doxorubicin-dependent ROS may explain, in part, Ca2+ and Na+ overload that contributes to diastolic dysfunction and development of heart failure. Therefore, we tested whether the administration of ranolazine, a selective blocker of late Na+ current, immediately after completing doxorubicin therapy, could affect diastolic dysfunction and interfere with the progression of functional decline. EXPERIMENTAL APPROACH: Fischer 344 rats received a cumulative dose of doxorubicin of 15 mg·kg-1 over a period of 2 weeks. After the assessment of diastolic dysfunction, the animals were treated with ranolazine (80 mg·kg-1 , daily) for the following 4 weeks. KEY RESULTS: While diastolic and systolic function progressively deteriorated in doxorubicin-treated animals, treatment with ranolazine relieved diastolic dysfunction and prevented worsening of systolic function, decreasing mortality. Ranolazine lowered myocardial NADPH oxidase 2 expression and oxidative/nitrative stress. Expression of the Na+ /Ca2+ exchanger 1 and Nav 1.5 channels was reduced and of the sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase 2 protein was increased. In addition, ranolazine lowered doxorubicin-induced hyper-phosphorylation and oxidation of Ca2+ /calmodulin-dependent protein kinase II, and decreased myocardial fibrosis. CONCLUSIONS AND IMPLICATIONS: Ranolazine, by the increased Na+ influx, induced by doxorubicin, altered cardiac Ca2+ and Na+ handling and attenuated diastolic dysfunction induced by doxorubicin, thus preventing the progression of cardiomyopathy. LINKED ARTICLES: This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.


Subject(s)
Doxorubicin/toxicity , Ranolazine/pharmacology , Sodium Channel Blockers/pharmacology , Ventricular Dysfunction, Left/prevention & control , Animals , Antibiotics, Antineoplastic/toxicity , Calcium/metabolism , Cardiotoxicity/etiology , Cardiotoxicity/prevention & control , Disease Models, Animal , Disease Progression , Female , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Rats , Rats, Inbred F344 , Sodium/metabolism , Ventricular Dysfunction, Left/chemically induced
8.
Br J Pharmacol ; 174(22): 4070-4086, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27922176

ABSTRACT

BACKGROUND AND PURPOSE: Heart failure with preserved ejection fraction (HFpEF) is a systemic syndrome driven by co-morbidities, and its pathophysiology is poorly understood. Several studies suggesting that dipeptidyl peptidase 4 (DPP4) might be involved in the pathophysiology of heart failure have prompted experimental and clinical investigations of DPP4 inhibitors in the cardiovascular system. Here we have investigated whether the DPP4 inhibitor sitagliptin affected the progression of HFpEF independently of its effects on glycaemia. EXPERIMENTAL APPROACH: Seven-week-old Dahl salt-sensitive rats were fed a high-salt diet for 5 weeks to induce hypertension. Then the rats continued with the high-salt diet and were treated with either sitagliptin (10 mg·kg-1 ) or vehicle for the following 8 weeks. Blood pressure and cardiac function were measured in vivo. Histochemical and molecular biology analyses of myocardium were used to assay cytokines, fibrotic markers, DPP4 and glucagon-like peptide-1 (GLP-1)/GLP-1 receptor. KEY RESULTS: Treatment with sitagliptin attenuated diastolic dysfunction, reduced mortality and reduced cardiac DPP4 activity, along with increased circulating GLP-1 and myocardial expression of GLP-1 receptors. Myocardial levels of pro-inflammatory cytokines (TNF-α, IL-6 and CCL2) were reduced. Sitagliptin treatment decreased the levels of endothelial NOS monomer, responsible for generation of ROS, while the amount of NO-producing dimeric form increased. Markers of oxidative and nitrosative stress were decreased. Moreover, increased collagen deposition and activation of pro-fibrotic signalling, inducing elevated myocardial stiffness, were attenuated by sitagliptin treatment. CONCLUSIONS AND IMPLICATIONS: Sitagliptin positively modulated active relaxation and passive diastolic compliance by decreasing inflammation-related endothelial dysfunction and fibrosis, associated with HFpEF. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Heart Failure/drug therapy , Sitagliptin Phosphate/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Blood Pressure/drug effects , Diastole/drug effects , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fibrosis , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Heart/drug effects , Heart/physiology , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Male , Myocardium/pathology , Nitric Oxide/metabolism , Rats, Inbred Dahl , Sitagliptin Phosphate/pharmacology , Stroke Volume
9.
Oncotarget ; 7(38): 62312-62326, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27694688

ABSTRACT

New strategies to prevent and early detect the cardiotoxic effects of the anticancer drug doxorubicin (DOXO) are required. MicroRNAs emerged as potential diagnostic, therapeutic and prognostic approaches in cardiovascular diseases. MiR-34a has a role in cardiac dysfunction and ageing and is involved in several cellular processes associated with DOXO cardiotoxicity. Our in vitro and in vivo results indicated that after DOXO exposure the levels of miR-34a are enhanced in cardiac cells, including Cardiac Progenitor Cells (CPCs). Since one of the determining event responsible for the initiation and evolution of the DOXO toxicity arises at the level of the CPC compartment, we evaluated if miR-34a pharmacological inhibition in these cells ameliorates the detrimental aftermath of the drug. AntimiR-34a has beneficial consequences on vitality, proliferation, apoptosis and senescence of DOXO-treated rat CPC. These effects are mediated by an increase of prosurvival miR-34a targets Bcl-2 and SIRT1, accompanied by a decrease of acetylated-p53 and p16INK4a. Importantly, miR-34a silencing also reduces the release of this miRNA from DOXO-exposed rCPCs, decreasing its negative paracrine effects on other rat cardiac cells. In conclusion, the silencing of miR-34a could represent a future therapeutic option for cardioprotection in DOXO toxicity and at the same time, it could be considered as a circulating biomarker for anthracycline-induced cardiac damage.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiomyopathies/blood , Doxorubicin/adverse effects , Heart/drug effects , MicroRNAs/metabolism , Myoblasts, Cardiac/metabolism , Acetylation , Animals , Apoptosis/drug effects , Biomarkers/analysis , Biomarkers/metabolism , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Cardiotoxicity/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Endothelial Cells , Female , MicroRNAs/antagonists & inhibitors , MicroRNAs/blood , Myoblasts, Cardiac/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism
10.
PLoS One ; 11(7): e0158746, 2016.
Article in English | MEDLINE | ID: mdl-27434719

ABSTRACT

BACKGROUND: The need for new options for chronic lung diseases promotes the research on stem cells for lung repair. Bone marrow-derived mesenchymal stem cells (MSCs) can modulate lung inflammation, but the data on cellular processes involved in early airway remodeling and the potential involvement of neuropeptides are scarce. OBJECTIVES: To elucidate the mechanisms by which local administration of MSCs interferes with pathophysiological features of airway hyperresponsiveness in an animal model. METHODS: GFP-tagged mouse MSCs were intratracheally delivered in the ovalbumin mouse model with subsequent functional tests, the analysis of cytokine levels, neuropeptide expression and histological evaluation of MSCs fate and airway pathology. Additionally, MSCs were exposed to pro-inflammatory factors in vitro. RESULTS: Functional improvement was observed after MSC administration. Although MSCs did not adopt lung cell phenotypes, cell therapy positively affected airway remodeling reducing the hyperplastic phase of the gain in bronchial smooth muscle mass, decreasing the proliferation of epithelium in which mucus metaplasia was also lowered. Decrease of interleukin-4, interleukin-5, interleukin-13 and increase of interleukin-10 in bronchoalveolar lavage was also observed. Exposed to pro-inflammatory cytokines, MSCs upregulated indoleamine 2,3-dioxygenase. Moreover, asthma-related in vivo upregulation of pro-inflammatory neurokinin 1 and neurokinin 2 receptors was counteracted by MSCs that also determined a partial restoration of VIP, a neuropeptide with anti-inflammatory properties. CONCLUSION: Intratracheally administered MSCs positively modulate airway remodeling, reduce inflammation and improve function, demonstrating their ability to promote tissue homeostasis in the course of experimental allergic asthma. Because of a limited tissue retention, the functional impact of MSCs may be attributed to their immunomodulatory response combined with the interference of neuropeptide system activation and tissue remodeling.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Receptors, Neurokinin-1/immunology , Receptors, Neurokinin-2/immunology , Respiratory Hypersensitivity/therapy , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Intubation, Intratracheal , Lung/immunology , Lung/pathology , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , Ovalbumin , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-2/genetics , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology
11.
Int J Cardiol ; 217: 69-79, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27179211

ABSTRACT

BACKGROUND: To investigate the effects of chronic administration of ranolazine (RAN) on experimental model of heart failure with preserved ejection fraction. METHODS: Seven-weeks old Dahl salt-sensitive rats were fed a high salt diet for 5weeks to induce hypertension. Afterwards, rats continued with a high salt diet and were administered either with vehicle or RAN (20mg/kg/die, ip) for the following 8weeks. Control rats were maintained on a low salt diet. RESULTS: While systolic parameters were not altered, diastolic parameters were changed in high salt animals. Hemodynamic analysis showed a decreased dP/dt min, increased LVEDP, longer time constant and steeper slope of the end-diastolic pressure-volume relationship. Treatment with RAN attenuated these alterations and determined a reduction in mortality. Additionally, the magnitude of myocardial hypertrophy and activation of PI3K/Akt pathway were reduced. Alteration in diastolic compliance as a consequence of elevated myocardial stiffness was confirmed by an increase of collagen deposition and activation of pro-fibrotic TGF-ß/SMAD3/CTGF signaling. These effects were counteracted by RAN. High salt rats had a decrease in SERCA2 and an increase in Na(+)/Ca(2+) exchanger (NCX). Treatment with RAN reduced NCX expression and determined an increment of SERCA2. Moreover, the levels of nitrotyrosine and oxidized dyhydroethidium were higher in high salt rats. RAN induced a decrement of oxidative stress, supporting the concept that reduction in ROS may mediate beneficial effects. CONCLUSIONS: Our findings support the possibility that diastolic dysfunction can be attenuated by RAN, indicating its ability to affect active relaxation and passive diastolic compliance.


Subject(s)
Cardiovascular Agents/administration & dosage , Heart Failure/drug therapy , Ranolazine/administration & dosage , Stroke Volume/drug effects , Ventricular Remodeling/drug effects , Animals , Disease Models, Animal , Drug Administration Schedule , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred Dahl , Signal Transduction/drug effects , Treatment Outcome
12.
Int J Cardiol ; 205: 99-110, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26730840

ABSTRACT

BACKGROUND: Doxorubicin (DOXO) is an effective anti-neoplastic drug but its clinical benefits are hampered by cardiotoxicity. Oxidative stress, apoptosis and myocardial fibrosis mediate the anthracycline cardiomyopathy. ROS trigger TGF-ß pathway that activates cardiac fibroblasts promoting fibrosis. Myocardial stiffness contributes to diastolic dysfunction, less studied aspect of anthracycline cardiomyopathy. Considering the role of SIRT1 in the inhibition of the TGF-ß/SMAD3 pathway, resveratrol (RES), a SIRT1 activator, might improve cardiac function by interfering with the development of cardiac fibrosis in a model of DOXO-induced cardiomyopathy. METHODS: F344 rats received a cumulative dose of 15 mg/kg of DOXO in 2 weeks or DOXO+RES (DOXO and RES, 2.5mg/kg/day, concomitantly for 2 weeks and then RES alone for 1 more week). The effects of RES on cardiac fibroblasts were also tested in vitro. RESULTS: Along with systolic dysfunction, DOXO was also responsible of diastolic abnormalities. Myocardial stiffness correlated with fibroblast activation and collagen deposition. DOXO+RES co-treatment significantly improved ± dP/dt and, more interestingly, ameliorated end-diastolic pressure/volume relationship. Treatment with RES resulted in reduced fibrosis and fibroblast activation and, most importantly, the mortality rate was significantly reduced in DOXO+RES group. Fibroblasts isolated from DOXO+RES-treated rats, in which SIRT1 was upregulated, showed decreased levels of TGF-ß and pSMAD3/SMAD3 when compared to cells isolated from DOXO-exposed hearts. CONCLUSIONS: Our findings reveal a key role of SIRT1 in supporting animal survival and functional parameters of the heart. SIRT1 activation by interfering with fibrogenesis can improve relaxation properties of myocardium and attenuate myocardial remodeling related to chemotherapy.


Subject(s)
Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Diastole/drug effects , Doxorubicin/toxicity , Sirtuin 1/metabolism , Stilbenes/therapeutic use , Animals , Anthracyclines/toxicity , Antibiotics, Antineoplastic/toxicity , Cardiomyopathies/chemically induced , Cells, Cultured , Disease Models, Animal , Female , Fibrosis , Rats , Rats, Inbred F344 , Resveratrol , Stilbenes/pharmacology
13.
Mediators Inflamm ; 2016: 3917471, 2016.
Article in English | MEDLINE | ID: mdl-28090152

ABSTRACT

Structural changes contribute to airway hyperresponsiveness and airflow obstruction in asthma. Emerging evidence points to the involvement of c-kit+ cells in lung homeostasis, although their potential role in asthma is unknown. Our aim was to isolate c-kit+ cells from normal mouse lungs and to test whether these cells can interfere with hallmarks of asthma in an animal model. Adult mouse GFP-tagged c-kit+ cells, intratracheally delivered in the ovalbumin-induced airway hyperresponsiveness, positively affected airway remodeling and improved airway function. In bronchoalveolar lavage fluid of cell-treated animals, a reduction in the number of inflammatory cells and in IL-4, IL-5, and IL-13 release, along with an increase of IL-10, was observed. In MSC-treated mice, the macrophage polarization to M2-like subset may explain, at least in part, the increment in the level of anti-inflammatory cytokine IL-10. After in vitro stimulation of c-kit+ cells with proinflammatory cytokines, the indoleamine 2,3-dioxygenase and TGFß were upregulated. These data, together with the increased apoptosis of inflammatory cells in vivo, indicate that c-kit+ cells downregulate immune response in asthma by influencing local environment, possibly by cell-to-cell contact combined to paracrine action. In conclusion, intratracheally administered c-kit+ cells reduce inflammation, positively modulate airway remodeling, and improve function. These data document previously unrecognized properties of c-kit+ cells, able to impede pathophysiological features of experimental airway hyperresponsiveness.


Subject(s)
Bronchial Hyperreactivity/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Airway Remodeling , Animals , Asthma/immunology , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Green Fluorescent Proteins/chemistry , Homeostasis , Immune System , Inflammation , Interleukin-10/therapeutic use , Lung/pathology , Macrophages/cytology , Male , Mice , Mice, Inbred BALB C
14.
Cardiooncology ; 2(1): 2, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-33530140

ABSTRACT

The cardiotoxicity of doxorubicin is becoming an interdisciplinary point of interest given a growing population of cancer survivors. The complex and not completely understood pathogenesis of this complication makes difficult to design successful preventive or curative measures. Although cardiomyocyte has been considered a classical cellular target, other cells including various types of undifferentiated cells are involved in myocardial homeostasis. Such perspective may shed light on previously unrecognized aspects of cardiotoxicity and promote new experimental and clinical cardioprotective strategies. In this review, different cellular targets of doxorubicin are discussed with the focus on cardiac progenitor cells, oxidative stress, DNA damage, senescence and apoptosis all of which contribute to their compromised functional properties.

15.
Int J Cardiol ; 189: 30-44, 2015.
Article in English | MEDLINE | ID: mdl-25889431

ABSTRACT

BACKGROUND: The search for compounds able to counteract chemotherapy-induced heart failure is extremely important at the age of global cancer epidemic. The role of SIRT1 in the maintenance of progenitor cell homeostasis may contribute to its cardioprotective effects. SIRT1 activators, by preserving progenitor cells, could have a clinical relevance for the prevention of doxorubicin (DOXO)-cardiotoxicity. METHODS: To determine whether SIRT1 activator, resveratrol (RES), interferes with adverse effects of DOXO on cardiac progenitor cells (CPCs): 1) human CPCs (hCPCs) were exposed in vitro to DOXO or DOXO+RES and their regenerative potential was tested in vivo in an animal model of DOXO-induced heart failure; 2) the in vivo effects of DOXO+RES co-treatment on CPCs were studied in a rat model. RESULTS: In contrast to healthy cells, DOXO-exposed hCPCs were ineffective in a model of anthracycline cardiomyopathy. The in vitro activation of SIRT1 decreased p53 acetylation, overcame suppression of the IGF-1/Akt pro-survival and anti-apoptotic signaling, enhanced oxidative stress defense and prevented senescence and growth arrest of hCPCs. Priming with RES counterbalanced the onset of dysfunctional phenotype in DOXO-exposed hCPCs, partly restoring their ability to repair the damage with improvement in cardiac function and animal survival. The in vivo co-treatment DOXO+RES prevented the anthracycline-induced alterations in CPCs, partly preserving cardiac function. CONCLUSION: SIRT1 activation protects DOXO-exposed CPCs and re-establishes their proper function. Pharmacological intervention at the level of tissue-specific progenitor cells may provide cardiac benefits for the growing population of long-term cancer survivors that are at risk of chemotherapy-induced cardiovascular toxicity.


Subject(s)
Cardiomyopathies/pathology , Doxorubicin/toxicity , Sirtuin 1/metabolism , Stem Cells/drug effects , Stilbenes/pharmacology , Analysis of Variance , Animals , Apoptosis/drug effects , Blotting, Western , Cardiomyopathies/chemically induced , Cardiomyopathies/drug therapy , Cells, Cultured , Disease Models, Animal , Doxorubicin/pharmacology , Female , Humans , Immunohistochemistry , Myocytes, Cardiac/drug effects , Normal Distribution , Random Allocation , Rats , Rats, Inbred F344 , Resveratrol , Sirtuin 1/drug effects , Statistics, Nonparametric , Stem Cells/physiology
16.
Mol Pharmacol ; 84(4): 603-14, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23913256

ABSTRACT

It is known that glutamate (Glu), the major excitatory amino acid in the central nervous system, can be an essential source for cell energy metabolism. Here we investigated the role of the plasma membrane Na(+)/Ca(2+) exchanger (NCX) and the excitatory amino acid transporters (EAATs) in Glu uptake and recycling mechanisms leading to ATP synthesis. We used different cell lines, such as SH-SY5Y neuroblastoma, C6 glioma and H9c2 as neuronal, glial, and cardiac models, respectively. We first observed that Glu increased ATP production in SH-SY5Y and C6 cells. Pharmacological inhibition of either EAAT or NCX counteracted the Glu-induced ATP synthesis. Furthermore, Glu induced a plasma membrane depolarization and an intracellular Ca(2+) increase, and both responses were again abolished by EAAT and NCX blockers. In line with the hypothesis of a mutual interplay between the activities of EAAT and NCX, coimmunoprecipitation studies showed a physical interaction between them. We expanded our studies on EAAT/NCX interplay in the H9c2 cells. H9c2 expresses EAATs but lacks endogenous NCX1 expression. Glu failed to elicit any significant response in terms of ATP synthesis, cell depolarization, and Ca(2+) increase unless a functional NCX1 was introduced in H9c2 cells by stable transfection. Moreover, these responses were counteracted by EAAT and NCX blockers, as observed in SH-SY5Y and C6 cells. Collectively, these data suggest that plasma membrane EAAT and NCX are both involved in Glu-induced ATP synthesis, with NCX playing a pivotal role.


Subject(s)
Adenosine Triphosphate/biosynthesis , Cell Membrane/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/pharmacology , Sodium-Calcium Exchanger/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Heart/drug effects , Heart/physiology , Humans , Rats
17.
Basic Res Cardiol ; 108(2): 334, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23411815

ABSTRACT

The increasing population of cancer survivors faces considerable morbidity and mortality due to late effects of the antineoplastic therapy. Cardiotoxicity is a major limiting factor of therapy with doxorubicin (DOXO), the most effective anthracycline, and is characterized by a dilated cardiomyopathy that can develop even years after treatment. Studies in animals have proposed the cardiac progenitor cells (CPCs) as the cellular target responsible for DOXO-induced cardiomyopathy but the relevance of these observations to clinical settings is unknown. In this study, the analysis of the DOXO-induced cardiomyopathic human hearts showed that the majority of human CPCs (hCPCs) was senescent. In isolated hCPCs, DOXO triggered DNA damage response leading to apoptosis early after exposure, and telomere shortening and senescence at later time interval. Functional properties of hCPCs, such as migration and differentiation, were also negatively affected. Importantly, the differentiated progeny of DOXO-treated hCPCs prematurely expressed the senescence marker p16(INK4a). In conclusion, DOXO exposure severely affects the population of hCPCs and permanently impairs their function. Premature senescence of hCPCs and their progeny can be responsible for the decline in the regenerative capacity of the heart and may represent the cellular basis of DOXO-induced cardiomyopathy in humans.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiomyopathy, Dilated/chemically induced , Cellular Senescence/drug effects , Doxorubicin/adverse effects , Myoblasts, Cardiac/drug effects , Stem Cells/drug effects , Adult , Antibiotics, Antineoplastic/therapeutic use , Blotting, Western , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Death/drug effects , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , DNA Damage/drug effects , Doxorubicin/therapeutic use , Female , Fluorescent Antibody Technique, Indirect , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-kit/metabolism , Telomere Homeostasis , beta-Galactosidase/metabolism
18.
J Cell Mol Med ; 16(4): 936-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21790971

ABSTRACT

Although low-energy extracorporeal cardiac shock wave (ECSW) therapy represents an attractive non-invasive treatment option for ischaemic heart disease, the precise mechanisms of its action and influence on the cardiac tissue remain obscure. The goal of this study was to evaluate the effects of SW application on cardiac function and structure. Four-month-old Fisher 344 rats were subjected to ECSW therapy. Echocardiographic measurements of cardiac function were performed at baseline and at 1 and 3 months after treatment. Signs of inflammation, apoptosis and fibrosis were evaluated by immunohistochemistry in the control and treated hearts. ECSW application did not provoke arrhythmia or increase the troponin-I level. At all time points, the left ventricular ejection fraction and fractional shortening remained stable. Histological analysis revealed neither differences in the extracellular matrix collagen content nor the presence of fibrosis; similarly, there were no signs of inflammation. Moreover, a population of cardiac cells that responded eagerly to ECSW application in the adult heart was identified; c-kit-positive, Ki67-positive, orthochromatic cells, corresponding to cardiac primitive cells, were 2.65-fold more numerous in the treated myocardium. In conclusion, non-invasive ECSW therapy is a safe and effective way of activating cardiac stem cells and myocardial regeneration. Because many factors influence cellular turnover in the ischaemic myocardium during the course of ischaemic heart disease, cardiac remodelling, and heart failure progression, studies to identify the optimal treatment time are warranted.


Subject(s)
Myocardial Ischemia/therapy , Animals , Male , Myocardial Ischemia/physiopathology , Rats , Rats, Inbred F344 , Regeneration
19.
Cardiovasc Hematol Agents Med Chem ; 8(1): 55-75, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20210776

ABSTRACT

Cardiovascular diseases and cancer represent respectively the first and second cause of death in industrialized countries. These two conditions may become synergistic when cardiovascular complications of anti-cancer therapy are considered. More than 70% of childhood and 50% of adult cancer patients can be cured, however this important success obtained by the biological and medical research is obfuscated by emerging findings of early and late morbidity due to cardiovascular events. Although anthracyclines are effective drugs against cancer a dose-dependent cardiotoxic effects whose mechanism has not been elucidated resulting in failure of therapeutic interventions limit their use. Unexpectedly, tyrosine/kinase inhibitors (TKIs) aimed at molecularly interfering with oncogenic pathways, have been implicated in cardiac side effects. Possible explanations of this phenomenon have been ambiguous, further strengthening the need to deepen our understanding on the mechanism of cardiotoxicity. In addition to a detailed description of anthracyclines and TKIs-related cardiovascular effects, the present review highlights recent observations supporting the hypothesis that the cellular target of anthracyclines and TKIs may include myocardial compartments other than parenchymal cells. The demonstration that the adult mammalian heart possesses a cell turnover regulated by primitive cells suggests that this cell population may be implicated in the onset and development of cardiovascular effects of anti-cancer strategies. The possibility of preventing cardiotoxicity by preservation and/or expansion of the resident stem cell pool responsible for cardiac repair may open new therapeutic options to unravel an unsolved clinical issue.


Subject(s)
Anthracyclines/adverse effects , Antineoplastic Agents/adverse effects , Cardiotoxins/adverse effects , Cardiovascular Diseases/chemically induced , Protein Kinase Inhibitors/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Humans , Myocardium/cytology , Myocardium/pathology , Neoplasms/drug therapy , Stem Cells/drug effects
20.
Circulation ; 121(2): 276-92, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20038740

ABSTRACT

BACKGROUND: Anthracyclines are the most effective drugs available in the treatment of neoplastic diseases; however, they have profound consequences on the structure and function of the heart, which over time cause a cardiomyopathy that leads to congestive heart failure. METHODS AND RESULTS: Administration of doxorubicin in rats led to a dilated myopathy, heart failure, and death. To test whether the effects of doxorubicin on cardiac anatomy and function were mediated by alterations in cardiac progenitor cells (CPCs), these cells were exposed to the anthracycline, which increased the formation of reactive oxygen species and caused increases in DNA damage, expression of p53, telomere attrition, and apoptosis. Additionally, doxorubicin resulted in cell-cycle arrest at the G2/M transition, which led to a significant decrease in CPC growth. Doxorubicin elicited multiple molecular adaptations; the massive apoptotic death that occurred in CPCs in the presence of anthracycline imposed on the surviving CPC pool the activation of several pathways aimed at preservation of the primitive state, cell division, lineage differentiation, and repair of damaged DNA. To establish whether delivery of syngeneic progenitor cells opposed the progression of doxorubicin cardiotoxicity, enhanced green fluorescent protein-labeled CPCs were injected in the failing myocardium; this treatment promoted regeneration of cardiomyocytes and vascular structures, which improved ventricular performance and rate of animal survival. CONCLUSIONS: Our results raise the possibility that autologous CPCs can be obtained before antineoplastic drugs are given to cancer patients and subsequently administered to individuals who are particularly sensitive to the cardiotoxicity of these agents for prevention or management of heart failure.


Subject(s)
Anthracyclines/adverse effects , Cardiomyopathies/chemically induced , Cardiomyopathies/therapy , Regeneration , Stem Cell Transplantation , Stem Cells/drug effects , Animals , Cardiomyopathies/pathology , Cardiomyopathy, Dilated/chemically induced , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/therapy , Cell Count , Doxorubicin/adverse effects , Heart Failure/chemically induced , Heart Failure/pathology , Heart Failure/therapy , Humans , Myocytes, Cardiac , Neoplasms/complications , Neoplasms/drug therapy , Rats , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...