Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
2.
Cardiovasc Drugs Ther ; 37(1): 89-105, 2023 02.
Article in English | MEDLINE | ID: mdl-34515894

ABSTRACT

PURPOSE: There is an increasing body of evidence to show that impairment in mitochondrial dynamics including excessive fission and insufficient fusion has been observed in the pre-diabetic condition. In pre-diabetic rats with cardiac ischemia-reperfusion (I/R) injury, acute treatment with a mitochondria fission inhibitor (Mdivi-1) and a fusion promoter (M1) showed cardioprotection. However, the potential preventive effects of chronic Mdivi-1 and M1 treatment in a pre-diabetic model of cardiac I/R have never been elucidated. METHODS: Male Wistar rats (n = 40) were fed with a high-fat diet (HFD) for 12 weeks to induce prediabetes. Then, all pre-diabetic rats received the following treatments daily via intraperitoneal injection for 2 weeks: (1) HFDV (Vehicle, 0.1% DMSO); (2) HFMdivi1 (Mdivi-1 1.2 mg/kg); (3) HFM1 (M1 2 mg/kg); and (4) HFCom (Mdivi-1 + M1). At the end of treatment protocols, all rats underwent 30 min of coronary artery ligation followed by reperfusion for 120 min. RESULTS: Chronic Mdivi-1, M1, and the combined treatment showed markedly improved cardiac mitochondrial function and dynamic control, leading to a decrease in cardiac arrhythmias, myocardial cell death, and infarct size (49%, 42%, and 51% reduction for HFMdivi1, HFM1, and HFCom, respectively vs HFDV). All of these treatments improved cardiac function following cardiac I/R injury in pre-diabetic rats. CONCLUSION: Chronic inhibition of mitochondrial fission and promotion of fusion exerted cardioprevention in prediabetes with cardiac I/R injury through the relief of cardiac mitochondrial dysfunction and dynamic alterations, and reduction in myocardial infarction, thus improving cardiac function.


Subject(s)
Diabetes Mellitus, Experimental , Myocardial Reperfusion Injury , Prediabetic State , Rats , Male , Animals , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/metabolism , Rats, Wistar , Mitochondrial Dynamics , Prediabetic State/drug therapy , Diabetes Mellitus, Experimental/metabolism , Myocytes, Cardiac , Mitochondria/metabolism , Apoptosis
3.
J Gerontol A Biol Sci Med Sci ; 78(3): 384-391, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36148512

ABSTRACT

Mitochondrial dysfunction is a factor potentially contributing to the Aging process. However, evidence surrounding changes in mitochondrial function and aging is still limited; therefore, this study aimed to investigate further the association between them. Possible confounding factors were included in the statistical analysis to explore the possibility of any independent associations. One thousand seven hundred and sixty-nine participants (619 middle-aged adults [age < 65] and 1,150 older adults [age ≥ 65]) from the Electricity Generating Authority of Thailand were enrolled in the study. The clinical characteristics and medical history were collected. Peripheral blood mononuclear cells (PBMCs) were isolated from venous blood and used for analysis of mitochondrial function. Several parameters pertinent to mitochondrial respiration including non-mitochondrial respiration, basal respiration, maximal respiration, proton leak, and spare respiratory capacity were found to be two to three times lower in the mitochondria isolated from the cells of older adults. Interestingly, the mitochondrial ATP production was only slightly reduced, and the percentage of coupling efficiency of PBMC mitochondria was significantly higher in the older adult group. The mitochondrial mass and oxidative stress were significantly reduced in older adult participants; however, the ratio of oxidative stress to mass was significantly increased. The association of these parameters with age was still shown to be the same from the outcome of the multivariate analyses. The mitochondrial functions and mitochondrial mass in PBMCs were shown to decline in association with age. However, the upregulation of mitochondrial oxidative stress production and mitochondrial coupling efficiency might indicate a compensatory response in mitochondria during aging.


Subject(s)
Cell Respiration , Leukocytes, Mononuclear , Humans , Aged , Middle Aged , Leukocytes, Mononuclear/metabolism , Cell Respiration/physiology , Mitochondria/metabolism , Aging , Oxidative Stress
4.
Endocrinol Metab (Seoul) ; 37(4): 630-640, 2022 08.
Article in English | MEDLINE | ID: mdl-35927067

ABSTRACT

BACKGRUOUND: High cardiorespiratory fitness (CRF) protects against age-related diseases. However, the mechanisms mediating the protective effect of high intrinsic CRF against metabolic, cardiac, and brain impairments in non-obese versus obese conditions remain incompletely understood. We aimed to identify the mechanisms through which high intrinsic CRF protects against metabolic, cardiac, and brain impairments in non-obese versus obese untrained rats. METHODS: Seven-week-old male Wistar rats were divided into two groups (n=8 per group) to receive either a normal diet or a highfat diet (HFD). At weeks 12 and 28, CRF, carbohydrate and fatty acid oxidation, cardiac function, and metabolic parameters were evaluated. At week 28, behavior tests were performed. At the end of week 28, rats were euthanized to collect heart and brain samples for molecular studies. RESULTS: The obese rats exhibited higher values for aging-related parameters than the non-obese rats, indicating that they experienced obesity-induced premature aging. High baseline CRF levels were positively correlated with several favorable metabolic, cardiac, and brain parameters at follow-up. Specifically, the protective effects of high CRF against metabolic, cardiac, and brain impairments were mediated by the modulation of body weight and composition, the lipid profile, substrate oxidation, mitochondrial function, insulin signaling, autophagy, apoptosis, inflammation, oxidative stress, cardiac function, neurogenesis, blood-brain barrier, synaptic function, accumulation of Alzheimer's disease-related proteins, and cognition. Interestingly, this effect was more obvious in HFD-fed rats. CONCLUSION: The protective effect of high CRF is mediated by the modulation of several mechanisms. These effects exhibit greater efficacy under conditions of obesity-induced premature aging.


Subject(s)
Aging, Premature , Cardiorespiratory Fitness , Insulin Resistance , Aging, Premature/metabolism , Aging, Premature/prevention & control , Animals , Brain/metabolism , Diet, High-Fat/adverse effects , Male , Obesity , Rats , Rats, Wistar
5.
Endocr Connect ; 11(4)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35275091

ABSTRACT

Objectives: To evaluate the effect of metformin in improving platelet dysfunction in women with gestational diabetes mellitus (GDM). Patients and methods: A randomized controlled trial was conducted on pregnant women diagnosed with GDM. Singleton low-risk pregnancies meeting the inclusion criteria were randomly allocated at 27-31 weeks to receive metformin and placebo through the rest of pregnancy. Thirty-seven and 39 cases were recruited into the metformin group and the placebo group, respectively. MPVs, P-selectin, and 8-isoprostane levels were determined at the time of allocation and 6 weeks after treatment. Obstetric and neonatal outcomes were also assessed. Results: Most baseline characteristics of the two groups were comparable. The levels of P-selectin after 6 weeks of treatment were significantly higher in the metformin group (68.9 ± 14.4 vs 60.6 ± 11.3; P-value = 0.006), indicating more platelet activation. All of the obstetric and neonatal outcomes were comparable except that birth weight was significantly lower in the metformin group (3018 ± 364 g vs 3204 ± 393 g; P-value = 0.037). Conclusion: Metformin, in addition to diet and lifestyle modifications, does not improve or worsen oxidative stress and platelet dysfunction in women with GDM. Nevertheless, metformin significantly reduces fetal weight in women with GDM, theoretically preventing macrosomia.

6.
Antioxidants (Basel) ; 11(2)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35204301

ABSTRACT

Tourniquet (TQ) use during total knee arthroplasty (TKA) induces ischemia/reperfusion (I/R) injury, resulting in mitochondrial dysfunction. This study aims to determine the effects of coenzyme Q10 (CoQ10) and ischemic preconditioning (IPC), either alone or in combination, on I/R-induced mitochondrial respiration alteration in peripheral blood mononuclear cells (PBMCs) and pain following TKA. Forty-four patients were allocated into four groups: control, CoQ10, IPC, and CoQ10 + IPC. CoQ10 dose was 300 mg/day for 28 days. IPC protocol was three cycles of 5/5-min I/R time. Mitochondrial oxygen consumption rates (OCRs) of PBMCs were measured seven times, at baseline and during ischemic/reperfusion phases, with XFe 96 extracellular flux analyzer. Postoperative pain was assessed for 48 h. CoQ10 improved baseline mitochondrial uncoupling state; however, changes in OCRs during the early phase of I/R were not significantly different from the placebo. Compared to ischemic data, IPC transiently increased basal OCR and ATP production at 2 h after reperfusion. Clinically, CoQ10 significantly decreased pain scores and morphine requirements at 24 h. CoQ10 + IPC abolished analgesic effect of CoQ10 and mitochondrial protection of IPC. In TKA with TQ, IPC enhanced mitochondrial function by a transient increase in basal and ATP-linked respiration, and CoQ10 provides postoperative analgesic effect. Surprisingly, CoQ10 + IPC interferes with beneficial effects of each intervention.

7.
Nutr Neurosci ; 25(6): 1115-1127, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33151133

ABSTRACT

BACKGROUND: Prediabetes can be characterized as obesity with metabolic disturbance, leading to cognitive decline and brain pathologies. D-allulose administration in obese animals decreased metabolic disturbance. However, the comparative effects of D-allulose and metformin on cognition and brain functions in the diet-induced prediabetic condition are unclear. We assume that both D-allulose and metformin equally restore cognition and brain functions in prediabetic rats to an equal extent. MATERIALS AND METHODS: Fifty-six rats were randomly divided into two groups: a control and diet-induced prediabetic group which had received a normal diet (ND) and a high-fat diet (HFD) for 24 weeks, respectively. After dietary protocol had been followed for 12 weeks, ND rats were given solely drinking water daily for 12 weeks. HFD-prediabetic rats randomly received drinking water with either D-allulose (1.9 g/kg/day of D-allulose) or metformin (300 mg/kg/day of metformin) for 12 weeks. Following this, cognition and brain parameters were determined. RESULTS: Brain oxidative stress, mitochondrial dysfunction, microglial hyper-activation, apoptosis, brain insulin insensitivity, hippocampal synaptic dysfunction, and cognitive decline were observed in prediabetic rats. D-allulose and metformin equally attenuated brain oxidative stress, brain mitochondrial ROS production, hippocampal apoptosis, brain insulin insensitivity, hippocampal synaptic dysfunction, resulting in improved learning process in prediabetic rats. Metformin conferred greater advantage on the amelioration of brain mitochondrial dysfunction and brain microglial hyper-activation than D-allulose, resulting in improvement in both learning and memory processes in prediabetic rats. CONCLUSIONS: Not only metformin, but also D-allulose, has beneficial effects on the enhancement of brain function and cognition in prediabetic condition.


Subject(s)
Cognitive Dysfunction , Drinking Water , Insulin Resistance , Insulins , Metformin , Prediabetic State , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Diet, High-Fat/adverse effects , Fructose , Insulin Resistance/physiology , Metformin/pharmacology , Metformin/therapeutic use , Obesity/metabolism , Prediabetic State/drug therapy , Rats , Rats, Wistar
8.
Acta Pharmacol Sin ; 43(1): 26-38, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33712720

ABSTRACT

Mitochondria are extraordinarily dynamic organelles that have a variety of morphologies, the status of which are controlled by the opposing processes of fission and fusion. Our recent study shows that inhibition of excessive mitochondrial fission by Drp1 inhibitor (Mdivi-1) leads to a reduction in infarct size and left ventricular (LV) dysfunction following cardiac ischemia-reperfusion (I/R) injury in high fat-fed induced pre-diabetic rats. In the present study, we investigated the cardioprotective effects of a mitochondrial fusion promoter (M1) and a combined treatment (M1 and Mdivi-1) in pre-diabetic rats. Wistar rats were given a high-fat diet for 12 weeks to induce prediabetes. The rats then subjected to 30 min-coronary occlusions followed by reperfusion for 120 min. These rats were intravenously administered M1 (2 mg/kg) or M1 (2 mg/kg) combined with Mdivi-1 (1.2 mg/kg) prior to ischemia, during ischemia or at the onset of reperfusion. We showed that administration of M1 alone or in combination with Mdivi-1 prior to ischemia, during ischemia or at the onset of reperfusion all significantly attenuated cardiac mitochondrial ROS production, membrane depolarization, swelling and dynamic imbalance, leading to reduced arrhythmias and infarct size, resulting in improved LV function in pre-diabetic rats. In conclusion, the promotion of mitochondrial fusion at any time-points during cardiac I/R injury attenuated cardiac mitochondrial dysfunction and dynamic imbalance, leading to decreased infarct size and improved LV function in pre-diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Myocardial Reperfusion Injury/metabolism , Prediabetic State/metabolism , Animals , Diabetes Mellitus, Experimental/chemically induced , Diet, High-Fat/adverse effects , Dose-Response Relationship, Drug , Male , Mitochondrial Dynamics/drug effects , Molecular Structure , Myocardial Reperfusion Injury/chemically induced , Prediabetic State/chemically induced , Quinazolinones/pharmacology , Rats , Rats, Wistar , Structure-Activity Relationship
9.
Front Med (Lausanne) ; 9: 1015306, 2022.
Article in English | MEDLINE | ID: mdl-36660002

ABSTRACT

Objective: To compare the levels of Non-transferrin bound iron (NTBI) in fetuses with anemia, using Hb Bart's disease as a study model, and those in unaffected fetuses and to determine the association between fetal cardiac function and the levels of NTBI. Patients and methods: A prospective study was conducted on pregnancies at risk of fetal Hb Bart's disease. All fetuses underwent standard ultrasound examination at 18-22 weeks of gestation for fetal biometry, anomaly screening and fetal cardiac function. After that, 2 ml of fetal blood was taken by cordocentesis to measure NTBI by Labile Plasma Iron (LPI), serum iron, hemoglobin and hematocrit. The NTBI levels of both groups were compared and the correlation between NTBI and fetal cardiac function was determined. Results: A total of 50 fetuses, including 20 fetuses with Hb Bart's disease and 30 unaffected fetuses were recruited. There was a significant increase in the level of serum iron in the affected group (median: 22.7 vs. 9.7; p-value: 0.013) and also a significant increase in NTBI when compared with those of the unaffected fetuses (median 0.11 vs. 0.07; p-value: 0.046). In comparisons of fetal cardiac function, myocardial performance (Tei) index of both sides was significantly increased in the affected group (left Tei: p = 0.001, Right Tei: p = 0.008). Also, isovolumetric contraction time (ICT) was also significantly prolonged (left ICT: p = 0.00, right ICT: p = 0.000). Fetal LPI levels were significantly correlated inversely with fetal hemoglobin levels (p = 0.030) but not significantly correlated with the fetal serum iron levels (p = 0.138). Fetal LPI levels were also significantly correlated positively with myocardial performance index (Tei) of both sides (right Tei: R = 0.000, left Tei: R = 0.000) and right ICT (R = 0.013), but not significantly correlated with left ICT (R = 0.554). Conclusion: Anemia caused by fetal Hb Bart's disease in pre-hydropic stage is significantly associated with fetal cardiac dysfunction and increased fetal serum NTBI levels which are significantly correlated with worsening cardiac dysfunction. Nevertheless, based on the limitations of the present study, further studies including long-term data are required to support a role of fetal anemia as well as increased fetal serum NTBI levels in development of subsequent heart failure or cardiac compromise among the survivors, possibly predisposing to cardiovascular disease in adult life.

10.
J Endocrinol ; 251(1): 27-39, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34265741

ABSTRACT

High-fat diet (HFD) consumption induces prediabetes and left ventricular dysfunction through many pathways including cell death pathway like necroptosis. Although the benefit of necroptosis inhibitor (necrostatin-1 or Nec-1) in the brain of prediabetic rats was shown, the effects of Nec-1 on cardiac autonomic function, blood pressure, cardiac function, along with its mechanistic insight have not been investigated. Male Wistar rats were fed with either a normal diet (n = 8) or HFD (n = 24) for 12 weeks to induce prediabetes. Prediabetic rats were randomly assigned into three interventional groups (n = 8/group): (1) vehicle, (2) Nec-1 (1.65 mg/kg, sc injection), and (3) metformin (300 mg/kg, oral gavage feeding). Treatments lasted for 8 weeks. Normal saline was given to normal diet-fed rats and vehicle group. Metabolic parameters, cardiac function and biochemical parameters were assessed. Prediabetic rats exhibited peripheral metabolic impairment as indicated by increased body weight, hyperinsulinemia with euglycemia, and dyslipidemia. Prediabetic rats also had cardiac autonomic imbalance, high blood pressure, and cardiac dysfunction, together with cardiac mitochondrial dysfunction, mitochondrial dynamic imbalance, and increased necroptosis and apoptosis. Treatment with Nec-1 did not affect peripheral metabolic parameters, however, it effectively reduced cardiac autonomic imbalance, blood pressure, and cardiac dysfunction via reducing cardiac inflammation, necroptosis, mitochondrial dysfunction, and increased mitochondrial fusion. Treatment with metformin reduced peripheral metabolic impairment and cardiac dysfunction via decreased cardiac mitochondrial dysfunction, mitochondrial dynamic imbalance, and apoptosis. In summary, Nec-1 directly suppressed necroptosis, cardiac mitochondrial dysfunction, and increased mitochondrial fusion independent of peripheral metabolic function, leading to an improved cardiac function in prediabetic rats.


Subject(s)
Imidazoles/pharmacology , Indoles/pharmacology , Mitochondria, Heart/drug effects , Prediabetic State/chemically induced , Prediabetic State/complications , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/etiology , Animals , Diet, High-Fat/adverse effects , Imidazoles/therapeutic use , Indoles/therapeutic use , Insulin Resistance , Male , Metformin/pharmacology , Obesity/etiology , Random Allocation , Rats , Rats, Wistar
11.
Neurotherapeutics ; 18(3): 2107-2125, 2021 07.
Article in English | MEDLINE | ID: mdl-34312765

ABSTRACT

Although doxorubicin (Dox) is an effective chemotherapy medication used extensively in the treatment of breast cancer, it frequently causes debilitating neurological deficits known as chemobrain. Donepezil (DPZ), an acetylcholinesterase inhibitor, provides therapeutic benefits in various neuropathological conditions. However, comprehensive mechanistic insights regarding the neuroprotection of DPZ on cognition and brain pathologies in a Dox-induced chemobrain model remain obscure. Here, we demonstrated that Dox-treated rats manifested conspicuous cognitive deficits and developed chemobrain pathologies as indicated by brain inflammatory and oxidative insults, glial activation, defective mitochondrial homeostasis, increased potential lesions associated with Alzheimer's disease, disrupted neurogenesis, loss of dendritic spines, and ultimately neuronal death through both apoptosis and necroptosis. Intervention with DPZ co-treatment completely restored cognitive function by attenuating these pathological conditions induced by DOX. We also confirmed that DPZ treatment does not affect the anti-cancer efficacy of Dox in breast cancer cells. Together, our findings suggest that DPZ treatment confers potential neuroprotection against Dox-induced chemobrain.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Chemotherapy-Related Cognitive Impairment/prevention & control , Donepezil/therapeutic use , Doxorubicin/toxicity , Inflammation Mediators/antagonists & inhibitors , Oxidative Stress/drug effects , Animals , Chemotherapy-Related Cognitive Impairment/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Donepezil/pharmacology , Female , Humans , Inflammation Mediators/metabolism , MCF-7 Cells , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , Rats , Rats, Wistar , Treatment Outcome
12.
Mitochondrion ; 59: 175-183, 2021 07.
Article in English | MEDLINE | ID: mdl-34091078

ABSTRACT

We aimed to compare mitochondrial function, mitochondrial dynamics, apoptosis, and necroptosis between odontogenic cysts/tumors, including radicular cysts, dentigerous cysts, ameloblastoma, vs. dental follicles as control. We demonstrated that mitochondrial dysregulation and imbalanced mitochondrial dynamics were observed in ameloblastoma. Apoptosis was increased in dentigerous cysts, and ameloblastoma, while necroptosis was suppressed in ameloblastoma. Necroptosis in radicular cysts was higher than that of control, suggesting that the inflammation-associated cell death occurred in radicular cysts. Our findings suggest ameloblastoma exhibited mitochondrial dysfunction, decreased mitochondrial fusion, and potential apoptosis. Therefore, alleviating mitochondrial dysregulation and apoptosis may be novel-targeted therapy for odontogenic cysts and tumors.


Subject(s)
Ameloblastoma/pathology , Dentigerous Cyst/pathology , Mitochondria/metabolism , Radicular Cyst/pathology , Reactive Oxygen Species/metabolism , Adolescent , Adult , Aged , Ameloblastoma/metabolism , Case-Control Studies , Cell Death , Child , Cross-Sectional Studies , Dentigerous Cyst/metabolism , Female , Humans , Male , Middle Aged , Mitochondrial Dynamics , Necroptosis , Radicular Cyst/metabolism , Young Adult
13.
Biochem Pharmacol ; 188: 114587, 2021 06.
Article in English | MEDLINE | ID: mdl-33932471

ABSTRACT

The standard chemotherapy regimens of ovarian cancer are platinum-based chemotherapy (carboplatin and paclitaxel) and bevacizumab (BEV). However, the effects of BEV alone or combined with carboplatin and paclitaxel on mitochondrial dynamics, mitochondrial function, mitophagy, apoptosis, inflammation and vascular endothelial growth factor (VEGF) in human ovarian cancer mitochondria and cells have not yet been investigated. Therefore, we aimed to test the hypothesis that 1) platinum-based chemotherapy and BEV equally damage isolated mitochondria from human ovarian cancers, and ovarian cancer cells through inducing mitochondrial dynamics dysregulation, mitochondrial dysfunction, increased mitophagy and apoptosis, as well as altered inflammation and VEGF; and 2) combined therapies exert greater damage than monotherapy. Each isolated human ovarian cancer mitochondria (n = 16) or CaOV3 cells (n = 6) were treated with either platinum-based chemotherapy (carboplatin 10 µM and paclitaxel 5 µM), BEV (2 mg/mL) or combined platinum-based chemotherapy and BEV for 60 min or 24 h, respectively. Following the treatment, mitochondrial dynamics, mitochondrial function, mitophagy, apoptosis, cytotoxicity, inflammation and VEGF were determined. Platinum-based chemotherapy caused ovarian cancer mitochondria and cell damage through mitochondrial dysfunction, increased cell death with impairment of membrane integrity, and enhanced VEGF reduction, while BEV did not. BEV caused deterioration of ovarian cancer mitochondria and cells through mitochondrial-dependent apoptosis, but it had no effect on cell viability. Interestingly, combined platinum-based chemotherapy and BEV treatments had no addictive effects on all parameters except mitochondrial maximal respiration, when compared to monotherapy. Collectively, these findings suggest that platinum-based chemotherapy and BEV caused human ovarian cancer mitochondrial and cell damage through different mechanisms.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bevacizumab/administration & dosage , Carboplatin/administration & dosage , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Female , Humans , Middle Aged , Platinum/administration & dosage , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Cells, Cultured
14.
J Adv Res ; 29: 33-44, 2021 03.
Article in English | MEDLINE | ID: mdl-33842003

ABSTRACT

Introduction: Previous studies reported the beneficial effects of pretreatment with melatonin on the heart during cardiac ischemia/reperfusion (I/R) injury. However, the effects of melatonin given after cardiac ischemia, as well as its comparative temporal effects are unknown. These include pretreatment, during ischemia, and at the onset of reperfusion. Also, the association between melatonin receptors and cardiac arrhythmias, mitochondrial function and dynamics, autophagy, and mitophagy during cardiac I/R have not been investigated. Objectives: We tested two major hypotheses in this study. Firstly, the temporal effect of melatonin administration exerts different cardioprotective efficacy during cardiac I/R. Secondly, melatonin provides cardioprotective effects via MT2 activation, leading to improvement in cardiac mitochondrial function and dynamics, reduced excessive mitophagy and autophagy, and decreased cardiac arrhythmias, resulting in improved LV function. Methods: Male rats were subjected to cardiac I/R, and divided into 4 intervention groups: vehicle, pretreatment with melatonin, melatonin given during ischemia, and melatonin given at the onset of reperfusion. In addition, either a non-specific melatonin receptor (MT) blocker or specific MT2 blocker was given to rats. Results: Treatment with melatonin at all time points alleviated cardiac I/R injury to a similar extent, quantified by reduction in infarct size, arrhythmia score, LV dysfunction, cardiac mitochondrial dysfunction, imbalance of mitochondrial dynamics, excessive mitophagy, and a decreased Bax/Bcl2 ratio. In H9C2 cells, melatonin increased %cell viability by reducing mitochondrial dynamic imbalance and a decrease in Bax protein expression. The cardioprotective effects of melatonin were dependent on MT2 activation. Conclusion: Melatonin given before or after ischemia exerted equal levels of cardioprotection on the heart with I/R injury, and its beneficial effects on cardiac arrhythmias, cardiac mitochondrial function and dynamics were dependent upon the activation of MT2.


Subject(s)
Cardiotonic Agents/pharmacology , Melatonin/pharmacology , Myocardial Reperfusion Injury/drug therapy , Receptor, Melatonin, MT2/metabolism , Animals , Apoptosis/drug effects , Arrhythmias, Cardiac/drug therapy , Autophagy/drug effects , Cardiotonic Agents/metabolism , Cell Line , Cell Survival/drug effects , Male , Mitochondria, Heart/drug effects , Mitochondrial Dynamics/drug effects , Mitophagy/drug effects , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Rats , Rats, Wistar , Receptors, Melatonin/metabolism , Signal Transduction/drug effects , Ventricular Function, Left/drug effects
15.
Aging (Albany NY) ; 13(8): 10955-10972, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33861726

ABSTRACT

Currently, the prevalence of obesity in aging populations is fast growing worldwide. Aging induced by D-galactose (D-gal) is proven to cause the worsening of cardiac dysfunction in pre-diabetic rats via deteriorating cardiac mitochondrial function. Hyperbaric oxygen therapy (HBOT) has been shown to attenuate D-gal-induced cognitive deterioration through decreased inflammation and apoptosis. We tested the hypothesis that HBOT alleviates D-gal induced cardiac dysfunction via improving mitochondrial function in pre-diabetic rats. Wistar rats (n=56) were fed normal diet or high-fat diet for 12 weeks. For subsequent 8 weeks, they were subcutaneously injected either vehicle (0.9% normal saline) or D-gal (150mg/kg/day). Rats were randomly subdivided into 7 groups at week 21: sham-treated (normal diet fed rats with vehicle (NDV), high-fat diet fed rats with vehicle (HFV), normal diet fed rats with D-gal (NDDg), high-fat diet fed rats with D-gal (HFDg)) and HBOT-treated (HFV, NDDg, HFDg). Sham rats received ambient pressure of oxygen while HBOT-treated ones received 100% oxygen given once daily for 60 minutes at 2 atmosphere absolute. HBOT reduced metabolic impairments, mitochondrial dysfunction and increased autophagy, resulting in an improvement of cardiac function in aged pre-diabetic rats.


Subject(s)
Aging/metabolism , Cardiovascular Diseases/therapy , Hyperbaric Oxygenation , Obesity/complications , Prediabetic State/therapy , Aging/drug effects , Animals , Apoptosis , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Galactose/administration & dosage , Galactose/toxicity , Humans , Injections, Subcutaneous , Male , Mitochondria, Heart/pathology , Obesity/metabolism , Obesity/therapy , Oxidative Stress , Oxygen/administration & dosage , Prediabetic State/complications , Rats , Rats, Wistar
16.
Mech Ageing Dev ; 195: 111465, 2021 04.
Article in English | MEDLINE | ID: mdl-33662435

ABSTRACT

The population of obese-elderly has increased prominently around the world. Both aging and obesity are major factors of neurodegeneration. The present study hypothesizes that HBOT attenuates metabolic disturbance, cognitive decline, hippocampal pathologies in aging and aging-obese model. Sixty Wistar rats were separated into 2 groups to receive normal-diet (ND) or high-fat diet (HFD) for 22 weeks. At week 13, ND rats were divided into two subgroups to receive vehicle (0.9 % NSS, s.c) or d-gal (150 mg/kg/d, s.c) for total 10 weeks. HFD rats were injected only d-gal (150 mg/kg/d, s.c; HFDD) for total 10 weeks. At week 20, rats in each subgroup were given sham-treatment (1ATA, 80 L/min, 80 min/day), or HBOT (2ATA, pure O2, 250 L/min, 80 min/day) for 14 days. Novel object location test, metabolic parameters, and hippocampal pathologies were determined after HBOT. d-gal induced insulin resistance, increased oxidative stress, autophagy impairment, microglial hyperactivation, apoptosis, synaptic dysplasticity which resulted in cognitive impairment. d-gal-treated HFD-fed rats had the highest levels of oxidative stress, apoptosis, dendritic spine loss. HBOT attenuated insulin resistance, cognitive impairment, hippocampal aging and pathologies in both models. These findings suggest that HBOT restored insulin sensitivity, hippocampal functions, cognition in aging and aging-obese models.


Subject(s)
Aging/physiology , Cognition/physiology , Cognitive Dysfunction , Hippocampus , Hyperbaric Oxygenation/methods , Obesity , Animals , Apoptosis , Behavior, Animal/physiology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/therapy , Hippocampus/metabolism , Hippocampus/pathology , Neuronal Plasticity , Obesity/metabolism , Obesity/psychology , Oxidative Stress , Rats , Rats, Wistar , Treatment Outcome
17.
Biochim Biophys Acta Mol Basis Dis ; 1867(1): 165975, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32956775

ABSTRACT

Cardiac ischemia/reperfusion (I/R) injury induces brain pathology. Donepezil, a well-known acetylcholine esterase (AChE) inhibitor, has been proven to exert neuroprotective effects against several neurodegenerative diseases. However, the comprehensive mechanism regarding the therapeutic potential of donepezil on the brain under cardiac I/R injury remains obscure. Here, we hypothesized that treatment with donepezil ameliorates brain pathology following cardiac I/R injury by decreasing blood brain barrier (BBB) breakdown, oxidative stress, neuroinflammation, mitochondrial dysfunction, mitochondrial dynamics imbalance, microglial activation, amyloid-beta (Aß) accumulation, neuronal apoptosis, and dendritic spine loss. Forty-eight adult male Wistar rats were subjected to surgery for cardiac I/R injury. Then, rats were randomly divided into four groups to receive either (1) saline (vehicle group), donepezil 3 mg/kg via intravenously administered (2) before ischemia (pretreatment group), (3) during ischemia (ischemia group), or (4) at the onset of reperfusion (reperfusion group). At the end of cardiac I/R paradigm, the brains were evaluated for BBB breakdown, brain inflammation, oxidative stress, mitochondrial function, mitochondrial dynamics, microglial morphology, Aß production, neuronal apoptosis, and dendritic spine density. Administration of donepezil at all time points equally showed an attenuation of brain damage in response to cardiac I/R injury, as indicated by increased expression of BBB junction protein, reduced brain inflammation and oxidative stress, improved mitochondrial function and mitochondrial dynamics, and alleviated Aß accumulation and microglial activation, resulting in protection of neuronal apoptosis and preservation of dendritic spine number. These findings suggest that donepezil potentially protects brain pathology caused by cardiac I/R injury regardless the timing of treatment.


Subject(s)
Alzheimer Disease/drug therapy , Brain Injuries/drug therapy , Donepezil/pharmacology , Myocardial Reperfusion Injury/drug therapy , Neuroprotective Agents/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Disease Models, Animal , Humans , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Oxidative Stress/drug effects , Rats , Rats, Wistar
18.
J Endocrinol ; 248(1): 59-74, 2021 01.
Article in English | MEDLINE | ID: mdl-33112810

ABSTRACT

Exogenous treatment of a neurotensin receptor 1 (NTR1) agonist exerted the neuroprotection in an obese and Alzheimer's model. However, the effects of NTR1 modulation on peripheral/hippocampal impairment and cognitive deficit following sustained HFD consumption are poorly understood. Forty rats received a normal diet (ND) or HFD for 16 weeks. At week 13, the ND group received a vehicle (n = 8). Thirty-two HFD-fed group were randomized into four subgroups (n = 8/subgroup) with a vehicle, 1 mg/kg of NTR1 agonist, 1 mg/kg of NTR antagonist, and combined treatment (NTR1 agonist-NTR antagonist) for 2 weeks, s.c. injection. Then, the cognitive tests and peripheral/hippocampal parameters were determined. Our findings demonstrated that NTR1 activator reversed obesity and attenuated metabolic impairment in pre-diabetic rats. It also alleviated hippocampal pathologies and synaptic dysplasticity, leading to deceleration or prevention of cognitive impairment progression. Therefore, NTR1 activation would be a possible novel therapy to decelerate or prevent progression of neuropathology and cognitive impairment in the pre-diabetes.


Subject(s)
Adamantane/analogs & derivatives , Cognitive Dysfunction/drug therapy , Hippocampus/drug effects , Imidazoles/therapeutic use , Obesity/drug therapy , Oligopeptides/therapeutic use , Receptors, Neurotensin/agonists , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Cognitive Dysfunction/etiology , Diet, High-Fat , Drug Evaluation, Preclinical , Drug Therapy, Combination , Hippocampus/metabolism , Ileum/drug effects , Ileum/metabolism , Imidazoles/pharmacology , Insulin Resistance , Male , Neuronal Plasticity/drug effects , Obesity/complications , Oligopeptides/pharmacology , Oxidative Stress/drug effects , Prediabetic State/drug therapy , Prediabetic State/metabolism , Random Allocation , Rats, Wistar , Receptors, Neurotensin/antagonists & inhibitors , Receptors, Neurotensin/metabolism
19.
Eur J Nutr ; 60(4): 2047-2061, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33011844

ABSTRACT

PURPOSE: Obesity-induced insulin resistant is associated with cardiovascular diseases via impairing cardiac mitochondria. Recently, D-allulose could protect ß-islets and improve insulin resistance. However, the effects of D-allulose on the heart and cardiac mitochondrial function under obesity-induced insulin-resistant condition has not been investigated. In this study, we aimed to investigate the effects of D-allulose on metabolic parameters, cardiac function, heart rate variability (HRV), cardiac mitochondrial function, and apoptosis in the heart of obesity-induced insulin-resistant rats induced by chronic high fat diet consumption. METHODS: Male Wistar rats (n = 24) received a normal fat diet (ND) or high fat diet (HFD) for 12 weeks. Then, HFD group was randomly divided into three subgroups to receive (1) HFD with distilled water, (2) HFD with 3% D-allulose 1.9 g/ kg·BW/ day (HFR), and (3) HFD with metformin 300 mg/kg·BW/ day (HFM) by diluted in drinking water daily for 12 weeks. At week 24, proposed study parameters were investigated. RESULTS: Chronic HFD consumption induced obesity-induced insulin resistant in rats and high fat diet impaired cardiac function and HRV. HFR rats had improved insulin sensitivity as indicated by decreasing HOMA index, plasma insulin, whereas HFM decreased body weight, visceral fat, plasma cholesterol, and plasma LDL. HFR and HFM provided similar efficacy in improving HRV and attenuating cardiac mitochondrial dysfunction, leading to improved cardiac function. CONCLUSIONS: Even though this is the first investigation of the D-allulose impact on the heart with a relatively small sample size, it clearly demonstrated a beneficial effect on the heart. D-allulose exerted a therapeutic effect on metabolic parameters except for body weight and lipid profiles and provided cardioprotective effects similar to metformin via attenuating cardiac mitochondrial function in obesity-induced insulin-resistant rats.


Subject(s)
Insulin Resistance , Insulin , Animals , Diet, High-Fat/adverse effects , Fructose , Male , Mitochondria, Heart , Obesity , Rats , Rats, Wistar
20.
Transl Res ; 230: 82-97, 2021 04.
Article in English | MEDLINE | ID: mdl-33137536

ABSTRACT

Cardiac autonomic imbalance including sympathetic overactivity and diminished parasympathetic activity is associated with left ventricular (LV) dysfunction in cases of cardiac ischemia/reperfusion (I/R) injury. Electrical stimulation to increase vagal activity has been shown to reduce infarct size and decrease fatal arrhythmias in cardiac I/R injury. However, the benefits of a parasympathomimetic drug on the heart during I/R are unclear. We hypothesized that administration of donepezil provides cardioprotection in cardiac I/R injury via reducing cellular apoptosis, oxidative stress, mitochondrial dysfunction, mitochondrial dynamic imbalance, increasing autophagy, and mitophagy. Fifty-four male Wistar rats were randomly assigned into sham and I/R groups. Acute cardiac I/R injury was induced by 30-minutes left anterior descending (LAD) coronary artery occlusion followed by 120-minutes reperfusion. These rats with induced I/R injury were randomly assigned to be treated with either: (1) Saline (vehicle group) or donepezil 3 mg/kg via intravenous injection given (2) before ischemia, (3) during ischemia, or (4) at the onset of reperfusion. Rats with cardiac I/R injury showed an increase in infarct size and arrhythmia score, LV dysfunction, impaired mitochondrial dynamic balance, autophagy and mitophagy, mitochondrial dysfunction, and increased apoptosis. All the donepezil-treated rats, regardless of the time of administration, showed a similar reduction in these impairments, and rebalancing in cardiac mitochondrial dynamics, leading to reduced myocardial infarct size and arrhythmia, and improved LV function. These findings suggested that donepezil effectively protected the heart against I/R injury through cardiac mitochondrial protection regardless of the time of administration.


Subject(s)
Autophagy/drug effects , Donepezil/pharmacology , Mitochondria, Heart/metabolism , Mitophagy/drug effects , Myocardial Infarction/complications , Myocardial Reperfusion Injury/drug therapy , Animals , Autophagy/physiology , Cholinesterase Inhibitors/pharmacology , Male , Mitophagy/physiology , Random Allocation , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...