Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-37502936

RESUMO

Aims: Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by cerebral amyloid ß (Aß) deposition and tau pathology. The AD-mediated degeneration of the brain neuro-signaling pathways, together with a potential peripheral amyloid accumulation, may also result in the derangement of the peripheral nervous system, culminating in detrimental effects on other organs, including the heart. However, whether and how AD pathology modulates cardiac function, neurotrophins, innervation, and amyloidosis is still unknown. Here, we report for the first time that cardiac remodeling, amyloid deposition, and neuro-signaling dysregulation occur in the heart of Tg2576 mice, a widely used model of AD and cerebral amyloidosis. Methods ad Results: Echocardiographic analysis showed significant deterioration of left ventricle function, evidenced by a decline of both ejection fraction and fraction shortening percentage in 12-month-old Tg2576 mice compared to age-matched WT littermates. Tg2576 mice hearts exhibited an accumulation of amyloid aggregates, including Aß, an increase in interstitial fibrosis and severe cardiac nervous system dysfunction. The transgenic mice also showed a significant decrease in cardiac nerve fiber density, including both adrenergic and regenerating nerve endings. This myocardial denervation was accompanied by a robust reduction in NGF and BDNF protein expression as well as GAP-43 expression (regenerating fibers) in both the brain and heart of Tg2576 mice. Accordingly, cardiomyocytes and neuronal cells challenged with Aß oligomers showed significant downregulation of BDNF and GAP-43, indicating a causal effect of Aß on the loss of cardiac neurotrophic function. Conclusions: Overall, this study uncovers possible harmful effects of AD on the heart, revealing cardiac degeneration induced by Aß through fibrosis and neuro-signaling pathway deregulation for the first time in Tg2576 mice. Our data suggest that AD pathology can cause deleterious effects on the heart, and the peripheral neurotrophic pathway may represent a potential therapeutic target to limit these effects.

2.
Cells ; 12(12)2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37371041

RESUMO

The adult heart is made up of cardiomyocytes (CMs) that maintain pump function but are unable to divide and form new myocytes in response to myocardial injury. In contrast, the developmental cardiac tissue is made up of proliferative CMs that regenerate injured myocardium. In mammals, CMs during development are diploid and mononucleated. In response to cardiac maturation, CMs undergo polyploidization and binucleation associated with CM functional changes. The transition from mononucleation to binucleation coincides with unique metabolic changes and shift in energy generation. Recent studies provide evidence that metabolic reprogramming promotes CM cell cycle reentry and changes in ploidy and nucleation state in the heart that together enhances cardiac structure and function after injury. This review summarizes current literature regarding changes in CM ploidy and nucleation during development, maturation and in response to cardiac injury. Importantly, how metabolism affects CM fate transition between mononucleation and binucleation and its impact on cell cycle progression, proliferation and ability to regenerate the heart will be discussed.


Assuntos
Traumatismos Cardíacos , Miócitos Cardíacos , Animais , Adulto , Humanos , Miócitos Cardíacos/metabolismo , Miocárdio , Ploidias , Traumatismos Cardíacos/metabolismo , Ciclo Celular/genética , Mamíferos
3.
Circ Res ; 132(7): 867-881, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36884028

RESUMO

BACKGROUND: Loss of brain-derived neurotrophic factor (BDNF)/TrkB (tropomyosin kinase receptor B) signaling accounts for brain and cardiac disorders. In neurons, ß-adrenergic receptor stimulation enhances local BDNF expression. It is unclear if this occurs in a pathophysiological relevant manner in the heart, especially in the ß-adrenergic receptor-desensitized postischemic myocardium. Nor is it fully understood whether and how TrkB agonists counter chronic postischemic left ventricle (LV) decompensation, a significant unmet clinical milestone. METHODS: We conducted in vitro studies using neonatal rat and adult murine cardiomyocytes, SH-SY5Y neuronal cells, and umbilical vein endothelial cells. We assessed myocardial ischemia (MI) impact in wild type, ß3AR knockout, or myocyte-selective BDNF knockout (myoBDNF KO) mice in vivo (via coronary ligation [MI]) or in isolated hearts with global ischemia-reperfusion (I/R). RESULTS: In wild type hearts, BDNF levels rose early after MI (<24 hours), plummeting at 4 weeks when LV dysfunction, adrenergic denervation, and impaired angiogenesis ensued. The TrkB agonist, LM22A-4, countered all these adverse effects. Compared with wild type, isolated myoBDNF KO hearts displayed worse infarct size/LV dysfunction after I/R injury and modest benefits from LM22A-4. In vitro, LM22A-4 promoted neurite outgrowth and neovascularization, boosting myocyte function, effects reproduced by 7,8-dihydroxyflavone, a chemically unrelated TrkB agonist. Superfusing myocytes with the ß3AR-agonist, BRL-37344, increased myocyte BDNF content, while ß3AR signaling underscored BDNF generation/protection in post-MI hearts. Accordingly, the ß1AR blocker, metoprolol, via upregulated ß3ARs, improved chronic post-MI LV dysfunction, enriching the myocardium with BDNF. Last, BRL-37344-imparted benefits were nearly abolished in isolated I/R injured myoBDNF KO hearts. CONCLUSIONS: BDNF loss underscores chronic postischemic heart failure. TrkB agonists can improve ischemic LV dysfunction via replenished myocardial BDNF content. Direct cardiac ß3AR stimulation, or ß-blockers (via upregulated ß3AR), is another BDNF-based means to fend off chronic postischemic heart failure.


Assuntos
Insuficiência Cardíaca , Isquemia Miocárdica , Neuroblastoma , Disfunção Ventricular Esquerda , Ratos , Camundongos , Humanos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Endoteliais/metabolismo , Neuroblastoma/metabolismo , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Receptores Adrenérgicos beta/metabolismo
4.
Front Physiol ; 14: 1060666, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36798942

RESUMO

The heart is a functional syncytium controlled by a delicate and sophisticated balance ensured by the tight coordination of its several cell subpopulations. Accordingly, cardiomyocytes together with the surrounding microenvironment participate in the heart tissue homeostasis. In the right atrium, the sinoatrial nodal cells regulate the cardiac impulse propagation through cardiomyocytes, thus ensuring the maintenance of the electric network in the heart tissue. Notably, the central nervous system (CNS) modulates the cardiac rhythm through the two limbs of the autonomic nervous system (ANS): the parasympathetic and sympathetic compartments. The autonomic nervous system exerts non-voluntary effects on different peripheral organs. The main neuromodulator of the Sympathetic Nervous System (SNS) is norepinephrine, while the principal neurotransmitter of the Parasympathetic Nervous System (PNS) is acetylcholine. Through these two main neurohormones, the ANS can gradually regulate cardiac, vascular, visceral, and glandular functions by turning on one of its two branches (adrenergic and/or cholinergic), which exert opposite effects on targeted organs. Besides these neuromodulators, the cardiac nervous system is ruled by specific neuropeptides (neurotrophic factors) that help to preserve innervation homeostasis through the myocardial layers (from epicardium to endocardium). Interestingly, the dysregulation of this neuro-signaling pathway may expose the cardiac tissue to severe disorders of different etiology and nature. Specifically, a maladaptive remodeling of the cardiac nervous system may culminate in a progressive loss of neurotrophins, thus leading to severe myocardial denervation, as observed in different cardiometabolic and neurodegenerative diseases (myocardial infarction, heart failure, Alzheimer's disease). This review analyzes the current knowledge on the pathophysiological processes involved in cardiac nervous system impairment from the perspectives of both cardiac disorders and a widely diffused and devastating neurodegenerative disorder, Alzheimer's disease, proposing a relationship between neurodegeneration, loss of neurotrophic factors, and cardiac nervous system impairment. This overview is conducive to a more comprehensive understanding of the process of cardiac neuro-signaling dysfunction, while bringing to light potential therapeutic scenarios to correct or delay the adverse cardiovascular remodeling, thus improving the cardiac prognosis and quality of life in patients with heart or neurodegenerative disorders.

5.
Sci Adv ; 8(36): eabq4293, 2022 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-36070391

RESUMO

Inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6i) are standard first-line treatments for metastatic ER+ breast cancer. However, acquired resistance to CDK4/6i invariably develops, and the molecular phenotypes and exploitable vulnerabilities associated with resistance are not yet fully characterized. We developed a panel of CDK4/6i-resistant breast cancer cell lines and patient-derived organoids and demonstrate that a subset of resistant models accumulates mitotic segregation errors and micronuclei, displaying increased sensitivity to inhibitors of mitotic checkpoint regulators TTK and Aurora kinase A/B. RB1 loss, a well-recognized mechanism of CDK4/6i resistance, causes such mitotic defects and confers enhanced sensitivity to TTK inhibition. In these models, inhibition of TTK with CFI-402257 induces premature chromosome segregation, leading to excessive mitotic segregation errors, DNA damage, and cell death. These findings nominate the TTK inhibitor CFI-402257 as a therapeutic strategy for a defined subset of ER+ breast cancer patients who develop resistance to CDK4/6i.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Neoplasias , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética
6.
JCI Insight ; 7(15)2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35771638

RESUMO

Developmental cardiac tissue is regenerative while operating under low oxygen. After birth, ambient oxygen is associated with cardiomyocyte cell cycle exit and regeneration. Likewise, cardiac metabolism undergoes a shift with cardiac maturation. Whether there are common regulators of cardiomyocyte cell cycle linking metabolism to oxygen tension remains unknown. The objective of the study is to determine whether mitochondrial UCP2 is a metabolic oxygen sensor regulating cardiomyocyte cell cycle. Neonatal rat ventricular myocytes (NRVMs) under moderate hypoxia showed increased cell cycle activity and UCP2 expression. NRVMs exhibited a metabolic shift toward glycolysis, reducing citrate synthase, mtDNA, mitochondrial membrane potential (ΔΨm), and DNA damage/oxidative stress, while loss of UCP2 reversed this phenotype. Next, WT and mice from a global UCP2-KO mouse line (UCP2KO) kept under hypoxia for 4 weeks showed significant decline in cardiac function that was more pronounced in UCP2KO animals. Cardiomyocyte cell cycle activity was reduced, while fibrosis and DNA damage was significantly increased in UCP2KO animals compared with WT under hypoxia. Mechanistically, UCP2 increased acetyl-CoA levels and histone acetylation, and it altered chromatin modifiers linking metabolism to cardiomyocyte cell cycle under hypoxia. Here, we show a potentially novel role for mitochondrial UCP2 as an oxygen sensor regulating cardiomyocyte cell cycle activity, acetyl-CoA levels, and histone acetylation in response to moderate hypoxia.


Assuntos
Proteínas Mitocondriais , Miócitos Cardíacos , Acetilcoenzima A/metabolismo , Acetilação , Animais , Ciclo Celular , Histonas/metabolismo , Hipóxia/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Oxigênio/metabolismo , Ratos , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
7.
JACC Basic Transl Sci ; 7(4): 366-380, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35540100

RESUMO

GRK5's catalytic activity in regulating basal and stressed cardiac function has not been studied. Herein, we studied knock-in mice in which GRK5 was mutated to render it catalytically inactive (K215R). At baseline, GRK5-K215R mice showed a marked decline in cardiac function with increased apoptosis and fibrosis. In vitro, restriction of GRK5 inside the nucleus of cardiomyocytes resulted in enhanced cell death along with higher p53 levels. Moreover, in fibroblasts, we demonstrated that K215R mutation promoted the transition into myofibroblast phenotype. This study provides novel insight into the biological actions of GRK5, that are essential for its future targeting.

8.
J Geriatr Cardiol ; 18(7): 549-559, 2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34404991

RESUMO

BACKGROUND: Aging is a multifactorial process associated with an impairment of autonomic nervous system (ANS) function. Progressive ANS remodeling includes upregulation of expression of circulating catecholamines and depletion of cardiac autonomic nerve fibers, and it is responsible, in part, for the increased susceptibility to cardiac diseases observed in elderly subjects. Neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF), are involved in synaptogenesis and neurite outgrowth processes, supporting neuronal cell differentiation and maturation. However, whether and how these factors and their downstream signaling are involved in cardiac aging remains unclear. Here, we tested whether, in the aged heart, the overall extent of autonomic fibers is reduced, owing to lower production of trophic factors such as BDNF and NGF. METHODS: In vivo, we used young (age: 3 months; n = 10) and old (age: 24 months; n = 11) male Fisher rats, whereas, we used human neuroblastoma (SH-SY5Y) cells in vitro. RESULTS: Compared to the young rats, old rats displayed a marked reduction in the overall ANS fiber density, affecting both sympathetic and cholinergic compartments, as indicated by dopamine ß-hydroxylase (dßh) and vesicular acetylcholine transporter (VaChT) immunohistochemical staining. In addition, a marked downregulation of GAP-43 and BDNF protein was observed in the left ventricular lysates of old rats compared to those of young rats. Interestingly, we did not find any significant difference in cardiac NGF levels between the young and old groups. To further explore the impact of aging on ANS fibers, we treated SH-SY5Y cells in vitro with serum obtained from young and old rats. Sera from both groups induced a remarkable increase in neuronal sprouting, as evidenced by a crystal violet assay. However, this effect was blunted in cells cultured with old rat serum and was accompanied by a marked reduction in GAP-43 and BDNF protein levels. CONCLUSIONS: Our data indicate that physiological aging is associated with an impairment of ANS structure and function and that reduced BDNF levels are responsible, at least in part, for these phenomena.

9.
Cell Mol Neurobiol ; 41(8): 1727-1742, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32844322

RESUMO

The primary forms of cell death seen in ischemic stroke are of two major types: a necrotic/necroptotic form, and an apoptotic form that is frequently seen in penumbral regions of injury. Typically apoptotic versus necroptotic programmed cell death is described as competitive in nature, where necroptosis is often described as playing a backup role to apoptosis. In the present study, we examined the relationship between these two forms of cell death in a murine endothelin-1 model of ischemia-reperfusion injury in wildtype and caspase-3 null mice with and without addition of the pharmacologic RIPK1 phosphorylation inhibitor necrostatin-1. Analyses of ischemic brain injury were performed via both cellular and volumetric assessments, electron microscopy, TUNEL staining, activated caspase-3 and caspase-7 staining, as well as CD11b and F4/80 staining. Inhibition of caspase-3 or RIPK1 phosphorylation demonstrates significant neural protective effects which are non-additive and exhibit significant overlap in protected regions. Interestingly, morphologic analysis of the cortex demonstrates reduced apoptosis following RIPK1 inhibition. Consistent with this, RIPK1 inhibition reduces the levels of both caspase-3 and caspase-7 activation. Additionally, this protection appears independent of secondary inflammatory mediators. Together, these observations demonstrate that the necroptotic protein RIPK1 modifies caspase-3/-7 activity, ultimately resulting in decreased neuronal apoptosis. These findings thus modify the traditional exclusionary view of apoptotic/necroptotic signaling, revealing a new form of interaction between these dominant forms of cell death.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/induzido quimicamente , Isquemia Encefálica/patologia , Endotelina-1/toxicidade , Animais , Apoptose/fisiologia , Isquemia Encefálica/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/induzido quimicamente , Necrose/metabolismo , Necrose/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
10.
Front Pharmacol ; 10: 888, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31447681

RESUMO

Hyperaldosteronism alters cardiac function, inducing adverse left ventricle (LV) remodeling either via increased fibrosis deposition, mitochondrial dysfunction, or both. These harmful effects are due, at least in part, to the activation of the G protein-coupled receptor kinase 2 (GRK2). In this context, we have previously reported that this kinase dysregulates both ß-adrenergic receptor (ßAR) and insulin (Ins) signaling. Yet, whether aldosterone modulates cardiac Ins sensitivity and ßAR function remains untested. Nor is it clear whether GRK2 has a role in this modulation, downstream of aldosterone. Here, we show in vitro, in 3T3 cells, that aldosterone impaired insulin signaling, increasing the negative phosphorylation of insulin receptor substrate 1 (ser307pIRS1) and reducing the activity of Akt. Similarly, aldosterone prevented the activation of extracellular signal-regulated kinase (ERK) and the production of cyclic adenosine 3',5'-monophosphate (cAMP) in response to the ß1/ß2AR agonist, isoproterenol. Of note, all of these effects were sizably reduced in the presence of GRK2-inhibitor CMPD101. Next, in wild-type (WT) mice undergoing chronic infusion of aldosterone, we observed a marked GRK2 upregulation that was paralleled by a substantial ß1AR downregulation and augmented ser307pIRS1 levels. Importantly, in keeping with the current in vitro data, we found that aldosterone effects were wholly abolished in cardiac-specific GRK2-knockout mice. Finally, in WT mice that underwent 4-week myocardial infarction (MI), we observed a substantial deterioration of cardiac function and increased LV dilation and fibrosis deposition. At the molecular level, these effects were associated with a significant upregulation of cardiac GRK2 protein expression, along with a marked ß1AR downregulation and increased ser307pIRS1 levels. Treating MI mice with spironolactone prevented adverse aldosterone effects, blocking GRK2 upregulation, and thus leading to a marked reduction in cardiac ser307pIRS1 levels while rescuing ß1AR expression. Our study reveals that GRK2 activity is a critical player downstream of the aldosterone signaling pathway; therefore, inhibiting this kinase is an attractive strategy to prevent the cardiac structural disarray and dysfunction that accompany any clinical condition accompanied by hyperaldosteronism.

11.
Vitam Horm ; 109: 387-406, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30678865

RESUMO

Aldosterone (Aldo) has been intensively studied for years since its isolation by Simpson and Tait in the 1950s. Interestingly, although most early research around Aldo's actions focused primarily on its interaction with the kidney, it was soon evident that this hormone is able to exert unexplained extra-renal effects, on various target organs including the heart. Importantly, over the course of the last decade a number of studies in pre-clinical models (in vitro and in vivo) and in humans have clearly demonstrated that Aldo, following the interaction with its receptor, the mineralocorticoid receptor (MR), is able to activate specific intracellular genomic and non-genomic pathways thus, regulating the homeostasis of the entire cardiovascular system. In this regard, Aldo has been shown to influence the function and growth of different type of cells including cardiomyocytes, fibroblasts and vascular cells (vascular smooth muscle cells and endothelial cells). Importantly, when present at high levels, Aldo have been associated with the onset of disparate cardiovascular disorders including heart failure (HF). For this reason, several pre-clinical study and clinical trials have been designed in order to test the effects of Aldo antagonism in HF patients. In this chapter, we will guide the readers through the current knowledge around Aldo activity in cardiovascular system and the relevant therapies acting in blocking the noxious Aldo/MR signaling pathway.


Assuntos
Aldosterona/metabolismo , Miocárdio/patologia , Regulação da Expressão Gênica , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo
12.
Oxid Med Cell Longev ; 2018: 1204598, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30327709

RESUMO

The mineralocorticoid hormone aldosterone (Aldo) has been intensively studied for its ability to influence both the physiology and pathophysiology of the cardiovascular system. Indeed, although research on Aldo actions for decades has mainly focused on its effects in the kidney, several lines of evidence have now demonstrated that this hormone exerts disparate extrarenal adverse effects, especially in the circulatory system. Accordingly, in the last lusters, a number of studies in preclinical models (in vitro and in vivo) and in humans have established that Aldo, following the interaction with its receptor-the mineralocorticoid receptor (MR)-is able to activate specific intracellular genomic and nongenomic pathways, thus regulating the homeostasis of the cardiovascular system. Importantly, through this mechanism of action, this hormone becomes a crucial regulator of the function and growth of different types of cells, including fibroblasts, cardiomyocytes, and vascular cells. For this main reason, it is plausible that when Aldo is present at high levels in the blood, it profoundly modifies the physiology of these cells, therefore being at the foundation of several cardiovascular disorders, such as heart failure (HF). On these grounds, in this review, we will provide an updated account on the current knowledge concerning Aldo activity in the cardiovascular system and the most recent preclinical studies and clinical trials designed to test better approaches able to counter the hyperactivity of the Aldo/MR signaling pathway in the setting of cardiovascular diseases.


Assuntos
Aldosterona/metabolismo , Fenômenos Fisiológicos Cardiovasculares , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Receptores de Mineralocorticoides/metabolismo , Animais , Humanos
13.
Med Hypotheses ; 114: 35-39, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29602461

RESUMO

Left ventricular hypertrophy (LVH) is a well established cardiovascular risk factor, accounting for an increase in cardiovascular morbid-mortality, although how much the magnitude and the kind of LVH could affect cardiovascular outcomes is in large part unknown. We speculate that mild LVH in absence of left ventricular (LV) chamber dilation, could play a protective role towards functional capacity, clinical outcome, cardiovascular and total morbi-mortality in conditions in which LV systolic function is generally reduced. Accordingly to many epidemiological observations, the availability of extra-quote of systolic function could lead to a significative improvement in the final outcome of some kinds of heart patients, as those undergoing bypass-grafting, where the stress for heart and cardiovascular system is always high. We suppose that the functional reserve available for patients with LVH could make the difference with respect to other patients undergoing myocardial revascularization. Similarly, the availability of a contractile reserve warranted by LVH could ensure a little gain in the outcome for patients after other major cardiovascular events (such as myocardial infarction or other heart surgery as surgical valve replacement). However, our hypothesis only involves mild LVH without LV chamber dilation, that is the initial stage of "non-dilated concentric" LVH and "non-dilated eccentric" LVH according to the new four-tiered classification of LVH based on relative wall thickness and LV dilation. Support for our hypothesis derives from the well-known protective role of systolic function that is a major factor in almost all cardiovascular diseases, where LV ejection fraction (LVEF) has shown to significantly improve quality of life, as well as morbidity and mortality. The knowledge that mild LVH in absence of LV chamber dilation is not as harmful in such conditions as believed at present could make avoidable some drugs prescription in some stages of the disease. Furthermore, it may allow a better evaluation of the risk profile of patients with LVH undergoing some cardiovascular major events like bypass grafting, myocardial infarction or surgical heart valve replacement.


Assuntos
Doenças Cardiovasculares/diagnóstico por imagem , Derivação Cardíaca Esquerda , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Valvas Cardíacas/cirurgia , Ventrículos do Coração/diagnóstico por imagem , Humanos , Modelos Teóricos , Contração Miocárdica , Qualidade de Vida , Fatores de Risco , Sístole , Função Ventricular Esquerda
14.
Circ Heart Fail ; 11(3): e004153, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29535114

RESUMO

BACKGROUND: Caloric restriction (CR) has been described to have cardioprotective effects and improve functional outcomes in animal models and humans. Chronic ischemic heart failure (HF) is associated with reduced cardiac sympathetic innervation, dysfunctional ß-adrenergic receptor signaling, and decreased cardiac inotropic reserve. We tested the effects of a long-term CR diet, started late after myocardial infarction on cardiac function, sympathetic innervation, and ß-adrenergic receptor responsiveness in a rat model of postischemic HF. METHODS AND RESULTS: Adult male rats were randomly assigned to myocardial infarction or sham operation and 4 weeks later were further randomized to a 1-year CR or normal diet. One year of CR resulted in a significant reduction in body weight, heart weight, and heart weight/tibia length ratio when compared with normal diet in HF groups. At the end of the study period, echocardiography and histology revealed that HF animals under the CR diet had ameliorated left ventricular remodeling compared with HF rats fed with normal diet. Invasive hemodynamic showed a significant improvement of cardiac inotropic reserve in CR HF rats compared with HF-normal diet animals. Importantly, CR dietary regimen was associated with a significant increase of cardiac sympathetic innervation and with normalized cardiac ß-adrenergic receptor levels in HF rats when compared with HF rats on the standard diet. CONCLUSIONS: We demonstrate, for the first time, that chronic CR, when started after HF established, can ameliorate cardiac dysfunction and improve inotropic reserve. At the molecular level, we find that chronic CR diet significantly improves sympathetic cardiac innervation and ß-adrenergic receptor levels in failing myocardium.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Infarto do Miocárdio/fisiopatologia , Tempo , Remodelação Ventricular/fisiologia , Adrenérgicos , Animais , Restrição Calórica/métodos , Modelos Animais de Doenças , Coração/inervação , Hemodinâmica/fisiologia , Masculino , Contração Miocárdica/fisiologia , Ratos Sprague-Dawley , Função Ventricular Esquerda/fisiologia
15.
Sarcoidosis Vasc Diffuse Lung Dis ; 35(3): 245-251, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-32476909

RESUMO

Introduction: Interstitial lung diseases (ILDs) are a heterogeneous group of lung disorders characterized by dyspnea on exertion, exercise-induced hypoxemia and reduced exercise tolerance. There are some evidences that pulmonary rehabilitation (PR) successfully treats these manifestations. Objective: To identify if pulmonary rehabilitation can achieve a clinically significant improvement of functional exercise capacity measured by 6-minute walk test (6-MWT) and arterial blood gases analysis (ABG) in patients with ILDs. Methods: A retrospective secondary analysis of the patients' records from in-patients of a pulmonary rehabilitation center between 2012 and 2015. Profiles of 38 patients who had completed 4-6 PR weeks were included. 6-MWT distance, dyspnea and fatigue Borg score and ABG at baseline and the end of the pulmonary rehabilitation were compared. Results: There was a statistically and clinically significant improvement in 6-MWT distance after PR with a mean difference for change in distance walked of 68.5±54.2 m. The pre post PR variation of dyspnea and fatigue Borg score significantly improved (-2.3±3.7, and -1.7±2.9, always p≤0.001). Among the ABG parameters, only the resting PaO2 showed a significant improvement after PR (Delta PaO2= 4.6±8.5 mmHg, p=0.005). Conclusions: A 4-6 week of PR improves functional exercise capacity and hypoxemia in patients with ILDs. (Sarcoidosis Vasc Diffuse Lung Dis 2018; 35: 245-251).

16.
Front Pharmacol ; 8: 556, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28878674

RESUMO

The sphingosine kinases 1 and 2 (SphK1 and 2) catalyze the phosphorylation of the lipid, sphingosine, generating the signal transmitter, sphingosine 1-phosphate (S1P). The activation of such kinases and the subsequent S1P generation and secretion in the blood serum of mammals represent a major checkpoint in many cellular signaling cascades. In fact, activating the SphK/S1P system is critical for cell motility and proliferation, cytoskeletal organization, cell growth, survival, and response to stress. In the cardiovascular system, the physiological effects of S1P intervene through the binding and activation of a family of five highly selective G protein-coupled receptors, called S1PR1-5. Importantly, SphK/S1P signal is present on both vascular and myocardial cells. S1P is a well-recognized survival factor in many tissues. Therefore, it is not surprising that the last two decades have seen a flourishing of interest and investigative efforts directed to obtain additional mechanistic insights into the signaling, as well as the biological activity of this phospholipid, and of its receptors, especially in the cardiovascular system. Here, we will provide an up-to-date account on the structure and function of sphingosine kinases, discussing the generation, release, and function of S1P. Keeping the bull's eye on the cardiovascular system, we will review the structure and signaling cascades and biological actions emanating from the stimulation of different S1P receptors. We will end this article with a summary of the most recent, experimental and clinical observations targeting S1PRs and SphKs as possible new therapeutic avenues for cardiovascular disorders, such as heart failure.

17.
Circ Res ; 118(8): 1244-53, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26926470

RESUMO

RATIONALE: It has been reported that epicardial adipose tissue (EAT) may affect myocardial autonomic function. OBJECTIVE: The aim of this study was to explore the relationship between EAT and cardiac sympathetic nerve activity in patients with heart failure. METHODS AND RESULTS: In 110 patients with systolic heart failure, we evaluated the correlation between echocardiographic EAT thickness and cardiac adrenergic nerve activity assessed by (123)I-metaiodobenzylguanidine ((123)I-MIBG). The predictive value of EAT thickness on cardiac sympathetic denervation ((123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score) was tested in a multivariate analysis. Furthermore, catecholamine levels, catecholamine biosynthetic enzymes, and sympathetic nerve fibers were measured in EAT and subcutaneous adipose tissue biopsies obtained from patients with heart failure who underwent cardiac surgery. EAT thickness correlated with (123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score, but not with left ventricular ejection fraction. Moreover, EAT resulted as an independent predictor of (123)I-MIBG early and late heart:mediastinum ratio and single-photon emission computed tomography total defect score and showed a significant additive predictive value on (123)I-MIBG planar and single-photon emission computed tomography results over demographic and clinical data. Although no differences were found in sympathetic innervation between EAT and subcutaneous adipose tissue, EAT showed an enhanced adrenergic activity demonstrated by the increased catecholamine levels and expression of catecholamine biosynthetic enzymes. CONCLUSIONS: This study provides the first evidence of a direct correlation between increased EAT thickness and cardiac sympathetic denervation in heart failure.


Assuntos
Tecido Adiposo/inervação , Fibras Adrenérgicas/diagnóstico por imagem , Insuficiência Cardíaca/diagnóstico por imagem , Pericárdio/inervação , Tecido Adiposo/diagnóstico por imagem , Idoso , Tomografia Computadorizada por Emissão de Fóton Único de Sincronização Cardíaca/métodos , Ecocardiografia/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tamanho do Órgão , Pericárdio/diagnóstico por imagem
18.
Circulation ; 109(21): 2587-93, 2004 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15136501

RESUMO

BACKGROUND: In hypertension, reduced nitric oxide production and blunted endothelial vasorelaxation are observed. It was recently reported that AKT phosphorylates and activates endothelial nitric oxide synthase and that impaired kinase activity may be involved in endothelial dysfunction. METHODS AND RESULTS: To identify the physiological role of the kinase in normotensive Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR), we used adenoviral vectors to transfer the human AKT1 gene selectively to the common carotid endothelium. In vitro, endothelial vasorelaxations to acetylcholine, isoproterenol, and insulin were blunted in control carotids from SHR compared with WKY rats, and human AKT1 overexpression corrected these responses. Similarly, blood flow assessed in vivo by Doppler ultrasound was reduced in SHR compared with WKY carotids and normalized after AKT1 gene transfer. In primary cultured endothelial cells, we evaluated AKT phosphorylation, activity, and compartmentalization and observed a mislocalization of the kinase in SHR. CONCLUSIONS: We conclude that AKT participates in the settings of endothelial dysfunction in SHR rats by impaired membrane localization. Our data suggest that AKT is involved in endothelium dysfunction in hypertension.


Assuntos
Endotélio Vascular/fisiopatologia , Hipertensão/fisiopatologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Acetilcolina/farmacologia , Adenoviridae/genética , Animais , Aorta , Artéria Carótida Primitiva/diagnóstico por imagem , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/fisiopatologia , Membrana Celular/enzimologia , Células Cultivadas/enzimologia , Células Endoteliais/enzimologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacologia , Humanos , Hipertensão/enzimologia , Hipertensão/genética , Injeções Intra-Arteriais , Insulina/farmacologia , Isoproterenol/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Proteínas Recombinantes de Fusão/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Ultrassonografia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...