Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.205
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1328139, 2024.
Article in English | MEDLINE | ID: mdl-38742195

ABSTRACT

The topic of human circadian rhythms is not only attracting the attention of clinical researchers from various fields but also sparking a growing public interest. The circadian system comprises the central clock, located in the suprachiasmatic nucleus of the hypothalamus, and the peripheral clocks in various tissues that are interconnected; together they coordinate many daily activities, including sleep and wakefulness, physical activity, food intake, glucose sensitivity and cardiovascular functions. Disruption of circadian regulation seems to be associated with metabolic disorders (particularly impaired glucose tolerance) and cardiovascular disease. Previous clinical trials revealed that disturbance of the circadian system, specifically due to shift work, is associated with an increased risk of type 2 diabetes mellitus. This review is intended to provide clinicians who wish to implement knowledge of circadian disruption in diagnosis and strategies to avoid cardio-metabolic disease with a general overview of this topic.


Subject(s)
Cardiovascular Diseases , Circadian Rhythm , Metabolic Diseases , Humans , Circadian Rhythm/physiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Metabolic Diseases/physiopathology , Metabolic Diseases/metabolism , Metabolic Diseases/etiology , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/metabolism , Chronobiology Disorders/physiopathology , Chronobiology Disorders/complications
2.
Psychiatry Res ; 337: 115951, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735240

ABSTRACT

Isolation of rodents throughout adolescence is known to induce many behavioral abnormalities which resemble neuropsychiatric disorders. Separately, this paradigm has also been shown to induce long-term metabolic changes consistent with a pre-diabetic state. Here, we investigate changes in central serotonin (5-HT) and glucagon-like peptide 1 (GLP-1) neurobiology that dually accompany behavioral and metabolic outcomes following social isolation stress throughout adolescence. We find that adolescent-isolation mice exhibit elevated blood glucose levels, impaired peripheral insulin signaling, altered pancreatic function, and fattier body composition without changes in bodyweight. These mice further exhibited disruptions in sleep and enhanced nociception. Using bulk and spatial transcriptomic techniques, we observe broad changes in neural 5-HT, GLP-1, and appetitive circuits. We find 5-HT neurons of adolescent-isolation mice to be more excitable, transcribe fewer copies of Glp1r (mRNA; GLP-1 receptor), and demonstrate resistance to the inhibitory effects of the GLP-1R agonist semaglutide on action potential thresholds. Surprisingly, we find that administration of semaglutide, commonly prescribed to treat metabolic syndrome, induced deficits in social interaction in group-housed mice and rescued social deficits in isolated mice. Overall, we find that central 5-HT circuitry may simultaneously influence mental well-being and metabolic health in this model, via interactions with GLP-1 and proopiomelanocortin circuitry.


Subject(s)
Disease Models, Animal , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Serotonin , Social Isolation , Animals , Mice , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Male , Serotonin/metabolism , Mental Disorders/metabolism , Mental Disorders/drug therapy , Mice, Inbred C57BL , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , Blood Glucose/metabolism , Blood Glucose/drug effects
3.
Exp Eye Res ; 242: 109885, 2024 May.
Article in English | MEDLINE | ID: mdl-38574944

ABSTRACT

The retinal microcirculation system constitutes a unique terminal vessel bed of the systemic circulation, and its perfusion status is directly associated with the neural function of the retina. This vascular network, essential for nourishing various layers of the retina, comprises two primary microcirculation systems: the retinal microcirculation and the choroidal microcirculation, with each system supplying blood to distinct retinal layers and maintaining the associated neural function. The blood flow of those capillaries is regulated via different mechanisms. However, a range of internal and external factors can disrupt the normal architecture and blood flow within the retinal microcirculation, leading to several retinal pathologies, including diabetic retinopathy, macular edema, and vascular occlusions. Metabolic disturbances such as hyperglycemia, hypertension, and dyslipidemia are known to modify retinal microcirculation through various pathways. These alterations are observable in chronic metabolic conditions like diabetes, coronary artery disease, and cerebral microvascular disease due to advances in non-invasive or minimally invasive retinal imaging techniques. Thus, examination of the retinal microcirculation can provide insights into the progression of numerous chronic metabolic disorders. This review discusses the anatomy, physiology and pathophysiology of the retinal microvascular system, with a particular emphasis on the connections between retinal microcirculation and systemic circulation in both healthy states and in the context of prevalent chronic metabolic diseases.


Subject(s)
Metabolic Diseases , Microcirculation , Retinal Vessels , Humans , Microcirculation/physiology , Retinal Vessels/physiopathology , Metabolic Diseases/physiopathology , Retinal Diseases/physiopathology , Regional Blood Flow/physiology
5.
Nat Metab ; 5(4): 563-571, 2023 04.
Article in English | MEDLINE | ID: mdl-37100995

ABSTRACT

Cardiometabolic diseases are a major public-health concern owing to their increasing prevalence worldwide. These diseases are characterized by a high degree of interindividual variability with regards to symptoms, severity, complications and treatment responsiveness. Recent technological advances, and the growing availability of wearable and digital devices, are now making it feasible to profile individuals in ever-increasing depth. Such technologies are able to profile multiple health-related outcomes, including molecular, clinical and lifestyle changes. Nowadays, wearable devices allowing for continuous and longitudinal health screening outside the clinic can be used to monitor health and metabolic status from healthy individuals to patients at different stages of disease. Here we present an overview of the wearable and digital devices that are most relevant for cardiometabolic-disease-related readouts, and how the information collected from such devices could help deepen our understanding of metabolic diseases, improve their diagnosis, identify early disease markers and contribute to individualization of treatment and prevention plans.


Subject(s)
Metabolic Diseases , Monitoring, Physiologic , Wearable Electronic Devices , Humans , Cardiovascular System/physiopathology , Continuous Glucose Monitoring , Data Collection , Fitness Trackers , Life Style , Metabolic Diseases/diagnosis , Metabolic Diseases/physiopathology , Metabolic Diseases/therapy , Monitoring, Physiologic/instrumentation , Monitoring, Physiologic/methods , Polysomnography , Time Factors , Wearable Electronic Devices/trends
6.
Int J Mol Sci ; 23(3)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35163627

ABSTRACT

Adipokines are a growing group of peptide or protein hormones that play important roles in whole body metabolism and metabolic diseases. Sleep is an integral component of energy metabolism, and sleep disturbance has been implicated in a wide range of metabolic disorders. Accumulating evidence suggests that adipokines may play a role in mediating the close association between sleep disorders and systemic metabolic derangements. In this review, we briefly summarize a group of selected adipokines and their identified function in metabolism. Moreover, we provide a balanced overview of these adipokines and their roles in sleep physiology and sleep disorders from recent human and animal studies. These studies collectively demonstrate that the functions of adipokine in sleep physiology and disorders could be largely twofold: (1) adipokines have multifaceted roles in sleep physiology and sleep disorders, and (2) sleep disturbance can in turn affect adipokine functions that likely contribute to systemic metabolic derangements.


Subject(s)
Adipokines/metabolism , Metabolic Diseases/metabolism , Sleep Wake Disorders/metabolism , Adipokines/physiology , Animals , Humans , Metabolic Diseases/physiopathology , Sleep , Sleep Apnea, Obstructive , Sleep Wake Disorders/physiopathology
7.
Molecules ; 27(3)2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35164383

ABSTRACT

A sedentary lifestyle or lack of physical activity increases the risk of different diseases, including obesity, diabetes, heart diseases, certain types of cancers, and some neurological diseases. Physical exercise helps improve quality of life and reduces the risk of many diseases. Irisin, a hormone induced by exercise, is a fragmented product of FNDC5 (a cell membrane protein) and acts as a linkage between muscles and other tissues. Over the past decade, it has become clear that irisin is a molecular mimic of exercise and shows various beneficial effects, such as browning of adipocytes, modulation of metabolic processes, regulation of bone metabolism, and functioning of the nervous system. Irisin has a role in carcinogenesis; numerous studies have shown its impact on migration, invasion, and proliferation of cancer cells. The receptor of irisin is not completely known; however, in some tissues it probably acts via a specific class of integrin receptors. Here, we review research from the past decade that has identified irisin as a potential therapeutic agent in the prevention or treatment of various metabolic-related and other diseases. This article delineates structural and biochemical aspects of irisin and provides an insight into the role of irisin in different pathological conditions.


Subject(s)
Fibronectins/metabolism , Metabolic Diseases/metabolism , Neoplasms/metabolism , Animals , Carcinogenesis/metabolism , Exercise , Fibronectins/analysis , Humans , Metabolic Diseases/physiopathology , Models, Molecular , Neoplasms/physiopathology , Protein Conformation , Sedentary Behavior , Signal Transduction
8.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054847

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) represents an increasing cause of liver disease worldwide, mirroring the epidemics of obesity and metabolic syndrome. As there are still no licensed medications for treating the disease, there is an ongoing effort to elucidate the pathophysiology and to discover new treatment pathways. An increasing body of evidence has demonstrated a crosstalk between the gut and the liver, which plays a crucial role in the development and progression of liver disease. Among other intestinal factors, gut permeability represents an interesting factor at the interface of the gut-liver axis. In this narrative review, we summarise the evidence from human studies showing the association between increased gut permeability and NAFLD, as well as with type-2 diabetes and obesity. We also discuss the manipulation of the gut permeability as a potential therapeutical target in patients with NAFLD.


Subject(s)
Intestines/pathology , Intestines/physiopathology , Metabolic Diseases/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Humans , Metabolic Diseases/therapy , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/therapy , Permeability
9.
Adv Drug Deliv Rev ; 181: 114110, 2022 02.
Article in English | MEDLINE | ID: mdl-34995679

ABSTRACT

Despite the increasing prevalence of obesity and diabetes, there is no efficient treatment to combat these epidemics. The adipose organ is the main site for energy storage and plays a pivotal role in whole body lipid metabolism and energy homeostasis, including remodeling and dysfunction of adipocytes and adipose tissues in obesity and diabetes. Thus, restoring and balancing metabolic functions in the adipose organ is in demand. MiRNAs represent a novel class of drugs and drug targets, as they are heavily involved in the regulation of many cellular and metabolic processes and diseases, likewise in adipocytes. In this review, we summarize key regulatory activities of miRNAs in the adipose organ, discuss various miRNA replacement and inhibition strategies, promising delivery systems for miRNAs and reflect the future of novel miRNA-based therapeutics to target adipose tissues with the ultimate goal to combat metabolic disorders.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/metabolism , Drug Delivery Systems/methods , Metabolic Diseases/physiopathology , MicroRNAs/pharmacology , Adipocytes/metabolism , Diabetes Mellitus, Type 2/physiopathology , Humans , Insulin Resistance/physiology , Lipid Metabolism/physiology , MicroRNAs/administration & dosage
10.
Hypertension ; 79(3): 518-531, 2022 03.
Article in English | MEDLINE | ID: mdl-34872333

ABSTRACT

BACKGROUND: Early-life exposures play key roles in the development of metabolic diseases. Whether such effects exist beyond one generation remains unclear. This study aimed to determine the transgenerational association of early-life exposure to the Chinese famine of 1959 to 1962 with the trajectories of body mass index (BMI), waist circumference (WC), and blood pressure (BP) in 2 consecutive generations. METHODS: We included 21 106 F1 observations born between 1954 and 1967 (median age: 45 years) and 1926 F2 observations (median age: 23 years) from the longitudinal household-based China Health and Nutrition Survey from 1993 to 2015. Trajectories of BMI, WC, systolic BP, and diastolic BP were fitted and compared between groups using linear mixed effect models. RESULTS: Early-life exposure to famine was associated with increased BMI, WC, and BP in 2 consecutive generations with sex and age disparities. In F1, famine was associated with increased BMI, WC, systolic BP, and diastolic BP, especially in men or those aged older than 50 years (P ranged from <0.001 to 0.02). In F2 men but not women, the parental exposure to famine was associated with 0.59 kg/m2 ([95% CI, 0.10-1.08], P=0.02) increase in BMI. In F2 aged ≥25 years but not those younger ones, the parental exposure to famine was associated with increased BMI (0.83 kg/m2 [0.14-1.51], P=0.03), systolic BP (2.04 mm Hg [0.20-3.88], P=0.03), and diastolic BP (1.73 mm Hg [0.28-3.18], P=0.02). CONCLUSIONS: The effects of an adverse developmental environment through famine in early life on BMI and BP later in life may persist beyond one generation.


Subject(s)
Blood Pressure/physiology , Body Mass Index , Famine , Metabolic Diseases/etiology , Waist Circumference/physiology , Adult , China , Female , Humans , Male , Metabolic Diseases/physiopathology , Middle Aged , Young Adult
11.
Shock ; 57(2): 238-245, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34678913

ABSTRACT

ABSTRACT: Persistent inflammation, immunosuppression, and catabolism syndrome (PIICS) is a growing challenge in intensive care units (ICUs). PIICS causes a severe illness with high mortality. Currently, treatment is expensive, and the outcomes are dismal. Herein, we established a PIICS model to study the disease pathophysiology and its potential treatment. Using a modified sublethal cecal ligation and puncture (CLP) to induce sepsis (day 1) and the injection of lipopolysaccharide (LPS) to induce an aggravated inflammation response (day 11), CLP + LPS mice recapitulating PIICS features were successfully generated (day 14). Adult male mice were divided into CLP + LPS, CLP + daily chronic stress (DCS), CLP, DCS, LPS, and sham control groups. A survival curve was generated, and phenotypes were analyzed using markers for catabolism, inflammation, and immunosuppression. The CLP + LPS model showed two mortality peaks (after CLP and after LPS), whereas the CLP + DCS and CLP groups showed one peak. Surviving CLP + LPS mice exhibited significantly increased catabolism and inflammatory cytokine levels and aggravated inflammation, including organ inflammation. CLP + LPS mice exhibited strong immune suppression as evidenced by decreased splenic cluster of differentiation (CD)8+ and interferon-γ+CD8+ T cell counts and a concomitant and significant increase in the myeloid-derived suppressor cell population. This CLP+LPS-induced PIICS model differs from acute sepsis models, showing two mortality peaks and a protracted course of 14 days. Compared to previous PIICS models, ours shows a re-aggravated status and higher catabolism, inflammation, and immunosuppression levels. Our aim was to use the PIICS model to simulate PIICS pathophysiology and course in the ICU, enabling investigation of its mechanism and treatment.


Subject(s)
Disease Models, Animal , Immune System Diseases/physiopathology , Immune System Diseases/therapy , Inflammation/physiopathology , Inflammation/therapy , Metabolic Diseases/physiopathology , Metabolic Diseases/therapy , Animals , Male , Mice , Syndrome
12.
Biomed Pharmacother ; 145: 112423, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34800783

ABSTRACT

Ferroptosis is a programmed iron-dependent cell death characterized by accumulation of lipid peroxides (LOOH) and redox disequilibrium. Ferroptosis shows unique characteristics in biology, chemistry, and gene levels, compared to other cell death forms. The metabolic disorder of intracellular LOOH catalyzed by iron causes the inactivity of GPX4, disrupts the redox balance, and triggers cell death. Metabolism of amino acid, iron, and lipid, including associated pathways, is considered as a specific hallmark of ferroptosis. Epidemiological studies and animal experiments have shown that ferroptosis plays an important character in the pathophysiology of cardiovascular disease such as atherosclerosis, myocardial infarction (MI), ischemia/reperfusion (I/R), heart failure (HF), cardiac hypertrophy, cardiomyopathy, and abdominal aortic aneurysm (AAA). This review systematically summarized the latest research progress on the mechanisms of ferroptosis. Then we report the contribution of ferroptosis in cardiovascular diseases. Finally, we discuss and analyze the therapeutic approaches targeting for ferroptosis associated with cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/physiopathology , Ferroptosis/physiology , Lipid Peroxides/metabolism , Animals , Cell Death/physiology , Humans , Metabolic Diseases/physiopathology , Oxidation-Reduction
13.
Biomed Pharmacother ; 146: 112582, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34959119

ABSTRACT

Growth and differentiation factor 15 (GDF-15) was discovered as a member of the transforming growth factor ß (TGF-ß) superfamily and the serum level of GDF-15 was significantly correlated with glucolipid metabolic disorders (GLMD) and cardiovascular diseases. In 2017, a novel identified receptor of GDF-15-glial-derived neurotrophic factor receptor alpha-like (GFRAL) was found to regulate energy homeostasis (such as obesity, diabetes and non-alcoholic fatty liver disease (NAFLD)). The function of GDF-15/GFRAL in suppressing appetite, enhancing glucose/lipid metabolism and vascular remodeling has been gradually revealed. These effects make it a potential therapeutic target for GLMD and vascular diseases. In this narrative review, we included and reviewed 121 articles by screening 524 articles from literature database. We primarily focused on the function of GDF-15 and its role in GLMD/cardiovascular diseases and discuss its potential clinical application.


Subject(s)
Cardiovascular Diseases/physiopathology , Growth Differentiation Factor 15/metabolism , Metabolic Diseases/physiopathology , Atherosclerosis/physiopathology , Clinical Trials as Topic , Diabetes Mellitus/physiopathology , Growth Differentiation Factor 15/chemistry , Humans , Hypertension/physiopathology , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/physiopathology
14.
Genes (Basel) ; 12(12)2021 12 09.
Article in English | MEDLINE | ID: mdl-34946914

ABSTRACT

Physical exercise has been associated with the modulation of micro RNAs (miRNAs), actively released in body fluids and recognized as accurate biomarkers. The aim of this study was to measure serum miRNA profiles in 18 horses taking part in endurance competitions, which represents a good model to test metabolic responses to moderate intensity prolonged efforts. Serum levels of miRNAs of eight horses that were eliminated due to metabolic unbalance (Non Performer-NP) were compared to those of 10 horses that finished an endurance competition in excellent metabolic condition (Performer-P). Circulating miRNA (ci-miRNA) profiles in serum were analyzed through sequencing, and differential gene expression analysis was assessed comparing NP versus P groups. Target and pathway analysis revealed the up regulation of a set of miRNAs (of mir-211 mir-451, mir-106b, mir-15b, mir-101-1, mir-18a, mir-20a) involved in the modulation of myogenesis, cardiac and skeletal muscle remodeling, angiogenesis, ventricular contractility, and in the regulation of gene expression. Our preliminary data open new scenarios in the definition of metabolic adaptations to the establishment of efficient training programs and the validation of athletes' elimination from competitions.


Subject(s)
Biomarkers/metabolism , Circulating MicroRNA/genetics , Gene Expression Regulation , Horses/physiology , Metabolic Diseases/physiopathology , Physical Conditioning, Animal , Transcriptome , Animals , Female , Male , Physical Endurance , Pilot Projects
15.
Int J Mol Sci ; 22(21)2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34768878

ABSTRACT

Many neurodegenerative and inherited metabolic diseases frequently compromise nervous system function, and mitochondrial dysfunction and oxidative stress have been implicated as key events leading to neurodegeneration. Mitochondria are essential for neuronal function; however, these organelles are major sources of endogenous reactive oxygen species and are vulnerable targets for oxidative stress-induced damage. The brain is very susceptible to oxidative damage due to its high metabolic demand and low antioxidant defence systems, therefore minimal imbalances in the redox state can result in an oxidative environment that favours tissue damage and activates neuroinflammatory processes. Mitochondrial-associated molecular pathways are often compromised in the pathophysiology of neurodegeneration, including the parkin/PINK1, Nrf2, PGC1α, and PPARγ pathways. Impairments to these signalling pathways consequently effect the removal of dysfunctional mitochondria, which has been suggested as contributing to the development of neurodegeneration. Mitochondrial dysfunction prevention has become an attractive therapeutic target, and there are several molecular pathways that can be pharmacologically targeted to remove damaged mitochondria by inducing mitochondrial biogenesis or mitophagy, as well as increasing the antioxidant capacity of the brain, in order to alleviate mitochondrial dysfunction and prevent the development and progression of neurodegeneration in these disorders. Compounds such as natural polyphenolic compounds, bioactive quinones, and Nrf2 activators have been reported in the literature as novel therapeutic candidates capable of targeting defective mitochondrial pathways in order to improve mitochondrial function and reduce the severity of neurodegeneration in these disorders.


Subject(s)
Metabolic Diseases/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Animals , Antioxidants/pharmacology , Humans , Metabolic Diseases/drug therapy , Metabolic Diseases/physiopathology , Mitochondria/drug effects , Mitochondria/pathology , Mitochondria/physiology , Mitophagy/drug effects , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neurons/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
16.
J Cardiovasc Pharmacol ; 78(Suppl 6): S78-S87, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34840260

ABSTRACT

ABSTRACT: Longer life span and increased prevalence of chronic, noncommunicable, inflammatory diseases fuel cardiovascular mortality. The microcirculation is central in the cross talk between ageing, inflammation, cardiovascular, and metabolic diseases. Microvascular dysfunction, characterized by alteration in the microvascular endothelial function and wall structure, is described in an increasing number of chronic age-associated diseases, suggesting that it might be a marker of ageing superior to chronological age. The aim of this review is to thoroughly explore the connections between microvascular dysfunction, ageing, and metabolic disorders by detailing the major role played by inflammation and oxidative stress in their evolution. Older age, hypertension, nutrient abundance, and hyperglycemia concur in the induction of a persistent low-grade inflammatory response, defined as meta-inflammation or inflammageing. This increases the local generation of reactive oxygen species that further impairs endothelial function and amplifies the local inflammatory response. Mitochondrial dysfunction is a hallmark of many age-related diseases. The alterations of mitochondrial function promote irreversible modification in microvascular structure. The interest in the hypothesis of chronic inflammation at the center of the ageing process lies in its therapeutic implications. Inhibition of specific inflammatory pathways has been shown to lower the risk of many age-related diseases, including cardiovascular disease. However, the whole architecture of the inflammatory response underpinning the ageing process and its impact on the burden of age-related diseases remain to be fully elucidated. Additional studies are needed to unravel the connection between these biological pathways and to address their therapeutic power in terms of cardiovascular prevention.


Subject(s)
Aging/metabolism , Cardiovascular Diseases/metabolism , Inflammation Mediators , Metabolic Diseases/metabolism , Microvessels/metabolism , Oxidative Stress , Age Factors , Aging/drug effects , Aging/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/prevention & control , Humans , Longevity , Metabolic Diseases/drug therapy , Metabolic Diseases/pathology , Metabolic Diseases/physiopathology , Microvessels/drug effects , Microvessels/pathology , Microvessels/physiopathology , Oxidative Stress/drug effects , Signal Transduction
17.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34596053

ABSTRACT

Circadian disruption is pervasive and can occur at multiple organizational levels, contributing to poor health outcomes at individual and population levels. Evidence points to a bidirectional relationship, in that circadian disruption increases disease severity and many diseases can disrupt circadian rhythms. Importantly, circadian disruption can increase the risk for the expression and development of neurologic, psychiatric, cardiometabolic, and immune disorders. Thus, harnessing the rich findings from preclinical and translational research in circadian biology to enhance health via circadian-based approaches represents a unique opportunity for personalized/precision medicine and overall societal well-being. In this Review, we discuss the implications of circadian disruption for human health using a bench-to-bedside approach. Evidence from preclinical and translational science is applied to a clinical and population-based approach. Given the broad implications of circadian regulation for human health, this Review focuses its discussion on selected examples in neurologic, psychiatric, metabolic, cardiovascular, allergic, and immunologic disorders that highlight the interrelatedness between circadian disruption and human disease and the potential of circadian-based interventions, such as bright light therapy and exogenous melatonin, as well as chronotherapy to improve and/or modify disease outcomes.


Subject(s)
Circadian Rhythm/physiology , Biomarkers , Cardiovascular Diseases/physiopathology , Humans , Mental Disorders/physiopathology , Mental Disorders/therapy , Metabolic Diseases/physiopathology , Neurodegenerative Diseases/physiopathology , Neurodevelopmental Disorders/physiopathology , Public Health
18.
J Cardiovasc Pharmacol ; 78(6): 858-866, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34596621

ABSTRACT

ABSTRACT: Paracetamol (PAR) is the most common over-the-counter drug recommended by physicians for treatment of pain and fever during gestation. This drug is not teratogenic, being considered safe for fetus; however, PAR crosses the blood-placental barrier. Considering that, the present study aimed to evaluate the vascular and metabolic safety of PAR exposure during intrauterine and neonatal development in adult male and female-exposed offspring. Wistar female rats were gavaged, with PAR (350 mg/kg/d), from gestational day 6-21 or from gestational day 6 until postnatal day 21. Control dams received water by gavage at the same periods. The male and female offspring were evaluated at adulthood (80 days of life). The thoracic aorta reactivity to acetylcholine, sodium nitroprusside, and phenylephrine was evaluated in male and female adult offspring. It was observed that aortic relaxation was similar between the PAR and control offspring. In addition, the contraction to phenylephrine was similar between the groups. Further, the insulin sensitivity, adipose tissue deposition and blood pressure were not different between PAR and control adult offspring. These results suggest that the protocol of PAR exposure used in the present study did not program vascular and metabolic alterations that would contribute to the development of cardiometabolic diseases in adult life, being safe for the exposed offspring.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Cardiovascular Diseases/chemically induced , Lactation , Metabolic Diseases/chemically induced , Prenatal Exposure Delayed Effects , Adiposity/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Cardiovascular Diseases/physiopathology , Female , Gestational Age , Hemodynamics/drug effects , Insulin Resistance , Male , Metabolic Diseases/blood , Metabolic Diseases/physiopathology , Pregnancy , Rats, Wistar , Risk Assessment
19.
Pharm Biol ; 59(1): 1161-1171, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34465259

ABSTRACT

CONTEXT: Various herbal medicines are thought to be useful in the management of cardiometabolic disease and its risk factors. Ganoderma lucidum (Curtis) P. Karst. (Ganodermataceae), also known as Lingzhi, has received considerable attention for various indications, including some related to the prevention and treatment of cardiovascular and metabolic disease by ameliorating major cardiovascular risk factors. OBJECTIVE: This review focuses on the major studies of the whole plant, plant extract, and specific active compounds isolated from G. lucidum in relation to the main risk factors for cardiometabolic disease. METHODS: References from major databases including PubMed, Web of Science, and Google Scholar were compiled. The search terms used were Ganoderma lucidum, Lingzhi, Reishi, cardiovascular, hypoglycaemic, diabetes, dyslipidaemia, antihypertensive, and anti-inflammatory. RESULTS: A number of in vitro studies and in vivo animal models have found that G. lucidum possesses antioxidative, antihypertensive, hypoglycaemic, lipid-lowering, and anti-inflammatory properties, but the health benefits in clinical trials are inconsistent. Among these potential health benefits, the most compelling evidence thus far is its hypoglycaemic effects in patients with type 2 diabetes or hyperglycaemia. CONCLUSIONS: The inconsistent evidence about the potential health benefits of G. lucidum is possibly because of the use of different Ganoderma formulations and different study populations. Further large controlled clinical studies are therefore needed to clarify the potential benefits of G. lucidum preparations standardised by known active components in the prevention and treatment of cardiometabolic disease.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Reishi/chemistry , Animals , Cardiometabolic Risk Factors , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Humans , Hypoglycemic Agents/pharmacology , Metabolic Diseases/drug therapy , Metabolic Diseases/physiopathology
20.
Bull Exp Biol Med ; 171(4): 431-434, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34542747

ABSTRACT

We studied the effect of the H2S donor (NaHS, 1-500 µM) on the contractile responses of isolated aortic smooth muscle segments from rats with metabolic syndrome induced by high-fat, high-carbohydrate diet. It was found that the vasorelaxing effect of NaHS (5-100 µM) decreased in under conditions of MS. The endothelial NO synthase inhibitor L-NAME (100 µM) suppressed the effect of NaHS, while cystathionine-gamma-lyase inhibitor PAG (100 µM) decreased the vasodilating effects of acetylcholine (0.1-100 µM). Application of endogenous NO precursor L-arginine (1 mM) potentiated in the effects of H2S donor NaHS. Thus, the contractile activity of vascular smooth muscles in metabolic syndrome is determined by not only the effect of H2S, but also the influence of NO.


Subject(s)
Hydrogen Sulfide/pharmacology , Metabolic Diseases/physiopathology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Animals , Biomechanical Phenomena/drug effects , Male , Metabolic Diseases/pathology , Metabolic Syndrome/pathology , Metabolic Syndrome/physiopathology , Muscle, Smooth, Vascular/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...