Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
FASEB J ; 37(11): e23262, 2023 11.
Article in English | MEDLINE | ID: mdl-37855727

ABSTRACT

Obesity accelerates the aging processes, resulting in an aggravation of aging-induced osteoporosis. We investigated the anti-osteoporotic effect of hyperbaric oxygen therapy (HBOT) in obese- and lean-aged rats through measurement of cellular senescence, hypoxia, inflammation, antioxidants, and bone microarchitecture. Obese and lean male Wistar rats were injected with 150 mg/kg/day of D-galactose for 8 weeks to induce aging. Then, all rats were randomly given either sham or HBOT for 14 days. Metabolic parameters were determined. Expression by bone mRNA for cellular senescence, hypoxia, inflammation, antioxidative capacity, and bone remodeling were examined. Micro-computed tomography and atomic absorption spectroscopy were performed to evaluate bone microarchitecture and bone mineral profiles, respectively. We found that HBOT restored the alterations in the mRNA expression level of p16, p21, HIF-1α, TNF-α, IL-6, RANKL, RANK, NFATc1, DC-STAMP, Osx, ALP, and Col1a1 in the bone in obese-and lean- aging rats. In obese-aging rats, HBOT increased the level of expression of Sirt1 and CuZnSOD mRNA and diminished the expression level of HIF-2α and ctsk mRNA to the same levels as the control group. However, HBOT failed to alter catalase and OCN mRNA expression in obese-aged rats. HBOT partially improved the bone microarchitecture in obese-aged rats, but completely restored it in lean-aged rats. Interestingly, HBOT protected against obesity-induced demineralization in obese-aged rats. In summary, HBOT exerts an anti-osteoporotic effect in lean-aged rats and prevents some, but not all the negative effects of obese-aged conditions on bone health. Therefore, HBOT is considered as a potential therapy for aging-induced osteoporosis, regardless of obese status.


Subject(s)
Hyperbaric Oxygenation , Osteoporosis , Rats , Male , Animals , Rats, Wistar , Galactose , X-Ray Microtomography , Obesity/complications , Obesity/therapy , Osteoporosis/etiology , Osteoporosis/therapy , Inflammation , Hypoxia , RNA, Messenger
2.
J Wound Care ; 32(10): 676-684, 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37830829

ABSTRACT

OBJECTIVE: This study aimed to investigate the involvement of mitochondrial biogenesis, and determine the extent of fibroblast proliferation and cellular apoptosis, in the gingiva of patients who had undergone head and neck radiation, after receiving hyperbaric oxygen therapy (HBOT), in comparison with normal gingiva. METHOD: A total of 16 patients who had undergone head and neck radiation with HBOT and six healthy subjects were included in the study. After the completion of radiation therapy, patients received HBOT at 2 ATA for 90 minutes per session, and for 20 sessions per patient. Samples of gingival tissues were then taken. The levels of: transforming growth factor beta (TGF-ß); phospho-nuclear factor kappa-light-chain-enhancer of activated B cells (p-NFÏ°B); nuclear factor kappa-light-chain-enhancer of activated B cells (NFÏ°B); proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α); phospho-dynamin-related protein 1 at ser616 (p-Drp1ser616); dynamin-related protein 1 (Drp1); Bcl-2-associated X-protein (Bax); and B-cell lymphoma 2 (Bcl-2) were determined using a Western blot. Independent t-test and Chi-squared tests were used in the study. RESULTS: There were no differences in the levels of TGF-ß, p-NFÏ°B, NFÏ°B, p-Drp1ser616, Drp1, Bax and Bcl-2 between the two groups. However, the level of PGC-1α was greater in irradiated gingival tissues with HBOT than in the healthy gingiva. CONCLUSION: Radiation-induced impaired wound healing can be improved by HBOT as indicated by levels of apoptosis, mitochondrial dynamics, cell proliferation and inflammation in irradiated gingiva with HBOT to a similar level to normal healthy gingiva. These findings may occur through an increase in mitochondrial biogenesis following HBOT.


Subject(s)
Hyperbaric Oxygenation , Humans , Gingiva , bcl-2-Associated X Protein , Wound Healing , Transforming Growth Factor beta , Dynamins
3.
Arch Biochem Biophys ; 740: 109598, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37054769

ABSTRACT

Inflammation and oxidative stress are mechanisms which potentially underlie the brain damage that can occur after cardiac ischemic and reperfusion (I/R) injury. 2i-10 is a new anti-inflammatory agent, acting via direct inhibition of myeloid differentiation factor 2 (MD2). However, the effects of 2i-10 and the antioxidant N-acetylcysteine (NAC) on pathologic brain in cardiac I/R injury are unknown. We hypothesized that 2i-10 and NAC offer similar neuroprotection levels against dendritic spine reduction through attenuation of brain inflammation, loss of tight junction integrity, mitochondrial dysfunction, reactive gliosis, and suppression of AD protein expression in rats with cardiac I/R injury. Male rats were allocated to either sham or acute cardiac I/R group (30 min of cardiac ischemia and 120 min of reperfusion). Rats in cardiac I/R group were given one of following treatments intravenously at the onset of reperfusion: vehicle, 2i-10 (20 or 40 mg/kg), and NAC (75 or 150 mg/kg). The brain was then used to determine biochemical parameters. Cardiac I/R led to cardiac dysfunction with dendritic spine loss, loss of tight junction integrity, brain inflammation, and mitochondrial dysfunction. Treatment with 2i-10 (both doses) effectively reduced cardiac dysfunction, tau hyperphosphorylation, brain inflammation, mitochondrial dysfunction, dendritic spine loss, and improved tight junction integrity. Although both doses of NAC effectively reduced brain mitochondrial dysfunction, treatment using a high dose of NAC reduced cardiac dysfunction, brain inflammation, and dendritic spine loss. In conclusion, treatment with 2i-10 and a high dose of NAC at the onset of reperfusion alleviated brain inflammation and mitochondrial dysfunction, consequently reducing dendritic spine loss in rats with cardiac I/R injury.


Subject(s)
Encephalitis , Myocardial Reperfusion Injury , Reperfusion Injury , Rats , Male , Animals , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Brain/metabolism , Oxidative Stress , Encephalitis/pathology , Ischemia/pathology
4.
Nutrients ; 14(22)2022 Nov 19.
Article in English | MEDLINE | ID: mdl-36432581

ABSTRACT

Interruptins A and B exhibited anti-diabetic, anti-inflammatory, and anti-oxidative effects. This study aimed to investigate the therapeutic ability of extract enriched by interruptins A and B (EEI) from an edible fern Cyclosorus terminans on insulin resistance and non-alcoholic fatty liver disease (NAFLD) in a high-fat diet (HFD)-induced obese rats and elucidate their possible mechanisms. HFD-induced obese rats were treated with EEI for 2 weeks. Real-time polymerase chain reaction (PCR) was used to examine the molecular basis. We found that EEI supplementation significantly attenuated body and liver weight gain, glucose intolerance, and insulin resistance. Concurrently, EEI increased liver and soleus muscle glycogen storage and serum high-density lipoprotein (HDL) levels. EEI also attenuated NAFLD, as indicated by improving liver function. These effects were associated with enhanced expression of insulin signaling genes (Slc2a2, Slc2a4, Irs1 and Irs2) along with diminished expression of inflammatory genes (Il6 and Tnf). Furthermore, EEI led to the suppression of lipogenesis genes, Srebf1 and Fasn, together with an increase in fatty acid oxidation genes, Ppara and Cpt2, in the liver. These findings suggest that EEI could ameliorate HFD-induced insulin resistance and NAFLD via improving insulin signaling pathways, inflammatory response, lipogenesis, and fatty acid oxidation.


Subject(s)
Insulin Resistance , Non-alcoholic Fatty Liver Disease , Tracheophyta , Rats , Animals , Diet, High-Fat/adverse effects , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/chemically induced , Insulin Resistance/genetics , Obesity/drug therapy , Obesity/etiology , Insulin/metabolism , Anti-Inflammatory Agents/pharmacology , Tracheophyta/metabolism , Fatty Acids/adverse effects
5.
Life Sci ; 295: 120406, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35182555

ABSTRACT

AIMS: To investigate the effects of hyperbaric oxygen therapy (HBOT) on metabolic disturbance, aging and bone remodeling in D-galactose-induced aging rats with and without obesity by determining the metabolic parameters, aging and oxidative stress markers, bone turnover markers, bone microarchitecture, and bone biomechanical strength. MATERIALS AND METHODS: Male Wistar rats were fed either a normal diet (ND; n = 18) or a HFD (n = 12) for 22 weeks. At week 13, vehicle (0.9% NaCl) was injected into ND-fed rats (NDV; n = 6), while 150 mg/kg/day of D-galactose was injected into 12 ND-fed rats (NDD) and 12 HFD-fed rats (HFDD) for 10 weeks. At week 21, rats were treated with either sham (NDVS, NDDS, or HFDDS; n = 6/ group) or HBOT (NDDH, or HFDDH; n = 6/group) for 14 days. Rats were then euthanized. Blood samples, femora, and tibiae were collected. KEY FINDINGS: Both NDD and HFDD groups developed aging as indicated by increased AGE level, increased inflammation and oxidative stress as shown by raised serum TNF-α and MDA levels, impaired bone remodeling as indicated by an increase in levels of CTX-1, TRACP-5b, and impaired bone structure/strength, when compared with those of the NDVS group. HFD aggravated these indicators of bone dyshomeostasis in D-galactose-treated rats. HBOT restored bone remodeling and bone structure/strength in the NDD group, however HBOT ameliorated bone dyshomeostasis in the HFDD group. SIGNIFICANCE: HBOT is a potential intervention to decrease the risk of osteoporosis and bone fracture in aging with or without obesity.


Subject(s)
Aging/physiology , Bone and Bones/metabolism , Hyperbaric Oxygenation/methods , Age Factors , Animals , Bone Remodeling/physiology , Bone and Bones/physiology , Diet, High-Fat , Galactose/adverse effects , Galactose/pharmacology , Homeostasis , Inflammation/metabolism , Insulin Resistance , Male , Obesity/metabolism , Obesity/physiopathology , Osteoporosis/metabolism , Oxidative Stress , Rats , Rats, Wistar
6.
Nutrients ; 13(9)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34579018

ABSTRACT

BACKGROUND: High-fat diet (HFD) consumption induced gut dysbiosis, inflammation, obese-insulin resistance. Perilla seed oil (PSO) is a rich source of omega-3 polyunsaturated fatty acids with health promotional effects. However, the effects of PSO on gut microbiota/inflammation and metabolic disturbance in HFD-induced obesity have not been investigated. Therefore, we aimed to compare the effects of different doses of PSO and metformin on gut microbiota/inflammation, and metabolic parameters in HFD-fed rats. METHODS: Thirty-six male Wistar rats were fed either a normal diet or an HFD for 24 weeks. At week 13, HFD-fed rats received either 50, 100, and 500 mg/kg/day of PSO or 300 mg/kg/day metformin for 12 weeks. After 24 weeks, the metabolic parameters, gut microbiota, gut barrier, inflammation, and oxidative stress were determined. RESULTS: HFD-fed rats showed gut dysbiosis, gut barrier disruption with inflammation, increased oxidative stress, metabolic endotoxemia, and insulin resistance. Treatment with PSO and metformin not only effectively attenuated gut dysbiosis, but also improved gut barrier integrity and decreased gut inflammation. PSO also decreased oxidative stress, metabolic endotoxemia, and insulin resistance in HFD-fed rats. Metformin had greater benefits than PSO. CONCLUSION: PSO and metformin had the beneficial effect on attenuating gut inflammation and metabolic disturbance in obese-insulin resistance.


Subject(s)
Dysbiosis/drug therapy , alpha-Linolenic Acid/therapeutic use , Animals , Blotting, Western , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/drug therapy , Insulin Resistance , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides/blood , Male , Metformin/therapeutic use , Oxidative Stress , Plant Oils/therapeutic use , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
7.
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
8.
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
9.
Am J Chin Med ; 49(2): 285-314, 2021.
Article in English | MEDLINE | ID: mdl-33622207

ABSTRACT

Irritable bowel syndrome (IBS) is a chronic dysfunction of the gastrointestinal tract, commonly characterized by abdominal pain or abdominal discomfort. These symptoms can substantially reduce the quality of life and work productivity of the patients. The exact pathogenesis of IBS remains unclear, as it has become apparent that multiple pathways are activated in the condition, including inflammation, immunology, neurology and psychology. Recent evidence has shown that symptoms in IBS are related to the dysfunction of the nervous system, particularly the viscerosomatic pathway, through immune-to-brain communication. The potential link between brain-gut relationships is gut microbiota. The management of IBS mostly focuses on symptomatically treating the patients. There are a wide range of standard treatments, including pharmacological to psychological interventions which are effective in some patients. Therefore, a combination of therapies including both standard and complimentary treatments, including Traditional Chinese Medicine (TCM) such as acupuncture, have been used in treating IBS patients. Several in vivo and clinical studies have demonstrated the efficacy of acupuncture in treating IBS. Increasing attention has been paid to research regarding the action mechanisms of acupuncture for IBS. This paper summarizes and discusses the possible mechanisms associated with acupuncture on the pathophysiology of IBS, including gastrointestinal (GI) motility, visceral hypersensitivity, the immune system, neurotransmitters, and the brain-gut axis. The results fromin vivo and clinical studies have been included. In addition, the effects of acupuncture on gut microbiota in IBS are included and any contradictory findings are deliberated.


Subject(s)
Acupuncture Therapy/methods , Gastrointestinal Microbiome/physiology , Irritable Bowel Syndrome/physiopathology , Irritable Bowel Syndrome/therapy , Acupuncture Points , Animals , Humans
10.
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
11.
Arch Osteoporos ; 15(1): 143, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32929613

ABSTRACT

INTRODUCTION: Iron overload, a state with excessive iron storage in the body, is a common complication in thalassemia patients which leads to multiple organ dysfunctions including the bone. Iron overload-induced bone disease is one of the most common and severe complications of thalassemia including osteoporosis. Currently, osteoporosis is still frequently found in thalassemia even with widely available iron chelation therapy. STUDY SELECTION: Relevant publications published before December 2019 in PubMed database were reviewed. Both pre-clinical studies and clinical trials were obtained using iron overload, thalassemia, osteoporosis, osteoblast, and osteoclast as keywords. RESULTS: Increased ROS production is a hallmark of iron overload-induced impaired bone remodeling. At the cellular level, oxidative stress affects bone remodeling by both osteoblast inhibition and osteoclast activation via many signaling pathways. In thalassemia patients, it has been shown that bone resorption was increased while bone formation was concurrently reduced. CONCLUSION: In this review, reports on the cellular mechanisms of iron overload-associated bone remodeling are comprehensively summarized and presented to provide current understanding this pathological condition. Moreover, current treatments and potential interventions for attenuating bone remodeling in iron overload are also summarized to pave ways for the future discoveries of novel agents that alleviate this condition.


Subject(s)
Bone Remodeling , Iron Overload/blood , Osteoporosis/etiology , Thalassemia/complications , Humans , Osteoclasts
12.
Eur J Nutr ; 59(4): 1453-1462, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31123863

ABSTRACT

PURPOSE: The chronic consumption of a high-fat diet (HFD) induces obese-insulin resistance and impairs jawbone health via gut dysbiosis-stimulated inflammatory process. Our previous studies demonstrated that the probiotic Lactobacillus paracasei HII01, prebiotic xylooligosaccharide (XOS), and synbiotics improved several vital organ functions by reducing gut dysbiosis in HFD-induced obese rats. However, the impacts on the cellular level of jawbone microarchitecture have not been examined. Here, we hypothesized that the supplementation of L. paracasei HII01, XOS, and synbiotics ameliorated the bone microarchitectural pathology in HFD-fed rats by reducing systemic inflammation and other metabolic parameters. METHODS: The dietary regimes (normal or high-fat diet) were provided to 48 male Wistar rats throughout 24-week experiment. After week 12, rats were given either a vehicle, pro-, pre-, or synbiotic for an additional 12 weeks before being killed. Then, blood analyses and bone histomorphometry of the jawbones were performed. RESULTS: The HFD-fed rats developed obese-insulin resistance with significantly elevated systemic inflammation. Bone histomorphometry of these rats showed a decrease in trabecular thickness with increased osteoclasts and active erosion surfaces. Mineral apposition and bone-formation rates were also remarkably diminished. The treatment with pro-, pre-, and synbiotics equally improved metabolic disturbance, reduced systemic inflammation, increased trabecular thickness, decreased osteoclasts and active erosion surfaces and restored mineral apposition and bone-formation rates. CONCLUSION: The probiotic L. paracasei HII01, prebiotic XOS, and the synbiotics had similarly beneficial effects to improve jawbone microarchitecture in HFD-fed rats by possibly ameliorating osteoclast-related bone resorption and potentiating bone-formation activities.


Subject(s)
Bone Diseases/prevention & control , Gastrointestinal Microbiome/drug effects , Inflammation/prevention & control , Lacticaseibacillus paracasei , Mandible/drug effects , Obesity/complications , Animals , Bone Diseases/etiology , Disease Models, Animal , Inflammation/etiology , Insulin Resistance , Male , Obesity/pathology , Rats , Rats, Wistar
13.
Toxicol Appl Pharmacol ; 382: 114741, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31473249

ABSTRACT

The present study aimed to compare the effects of high dose atorvastatin and a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor on the mitochondrial function in oxidative muscle fibers in obese female rats. Female Wistar rats were fed with either a normal diet (ND: n = 12) or a high-fat diet (HFD: n = 36) for a total of 15 weeks. At week 13, ND-fed rats received a vehicle, and HFD-fed rats were divided to three groups to receive either a vehicle, 40 mg/kg/day of atorvastatin, or 4 mg/kg/day of PCSK9 inhibitor (SBC-115076) for 3 weeks. Soleus muscles were investigated to assess mitochondrial ROS, membrane potential, swelling, mitochondrial-related protein expression, and level of malondialdehyde (MDA). The results showed that HFD-fed rats with vehicle developed obese-insulin resistance and dyslipidemia. Both atorvastatin and PCSK9 inhibitor reduced obesity and dyslipidemia, as well as improved insulin sensitivity in HFD-fed rats. However, the efficacy of PCSK9 inhibitor to increase weight loss and reduce dyslipidemia in HFD-fed rats was greater than those of atorvastatin. An increase in MDA level, ratio of p-Drp1ser616/total Drp1 protein, CPT1 protein, mitochondrial ROS, and membrane depolarization in the soleus muscle were observed in HFD-fed rats with vehicle. PCSK9 inhibitor enabled the restoration of all these parameters to normal levels. However, atorvastatin facilitated restoration of some parameters, including MDA level, p-Drp1ser616/total Drp1 ratio, and CPT1 protein expression. These findings suggest that PCSK9 inhibitor is superior to atorvastatin in instigating weight loss, cholesterol reduction, and attenuation of mitochondrial oxidative stress in oxidative muscle fibers of obese female rats.


Subject(s)
Atorvastatin/administration & dosage , Enzyme Inhibitors/administration & dosage , Insulin Resistance/physiology , Mitochondria/drug effects , Obesity/drug therapy , PCSK9 Inhibitors , Animals , Diet, High-Fat/adverse effects , Female , Mitochondria/metabolism , Obesity/etiology , Obesity/metabolism , Proprotein Convertase 9/metabolism , Rats , Rats, Wistar
14.
Free Radic Res ; 53(8): 901-909, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31387429

ABSTRACT

Although coenzyme Q10 (CoQ10) supplementation has shown to reduce pain levels in chronic pain, the effects of CoQ10 supplementation on pain, anxiety, brain activity, mitochondrial oxidative stress, antioxidants, and inflammation in pregabalin-treated fibromyalgia (FM) patients have not clearly elucidated. We hypothesised that CoQ10 supplementation reduced pain better than pregabalin alone via reducing brain activity, mitochondrial oxidative stress, inflammation, and increasing antioxidant levels in pregabalin-treated FM patients. A double-blind randomised placebo-controlled trial was conducted. Eleven FM patients were enrolled with 2 weeks wash-out then randomly allocated to 2 treatment groups; pregabalin with CoQ10 or pregabalin with placebo for 40 d. Then, patients in CoQ10 group were switched to placebo, and patients in placebo group were switched to CoQ10 for another 40 d. Pain pressure threshold (PPT), FM questionnaire, anxiety, and pain score were examined. Peripheral blood mononuclear cells (PBMCs) were isolated to investigate mitochondrial oxidative stress and inflammation at day 0, 40, and 80. The level of antioxidants and brain positron emission tomography (PET) scan were also determined at these time points. Pregabalin alone reduced pain and anxiety via decreasing brain activity compared with their baseline. However, it did not affect mitochondrial oxidative stress and inflammation. Supplementation with CoQ10 effectively reduced greater pain, anxiety and brain activity, mitochondrial oxidative stress, and inflammation. CoQ10 also increased a reduced glutathione levels and superoxide dismutase (SOD) levels in FM patients. These findings provide new evidence that CoQ10 supplementation provides further benefit for relieving pain sensation in pregabalin-treated FM patients, possibly via improving mitochondrial function, reducing inflammation, and decreasing brain activity.


Subject(s)
Fibromyalgia/drug therapy , Oxidative Stress , Pain/drug therapy , Pregabalin/therapeutic use , Ubiquinone/analogs & derivatives , Adult , Brain/diagnostic imaging , Brain/physiopathology , Double-Blind Method , Female , Fibromyalgia/complications , Fibromyalgia/diagnostic imaging , Fibromyalgia/physiopathology , Humans , Inflammation , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Mitochondria/metabolism , Pain/etiology , Positron-Emission Tomography , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
15.
Oxid Med Cell Longev ; 2018: 8087598, 2018.
Article in English | MEDLINE | ID: mdl-30524661

ABSTRACT

Ischemia and reperfusion (I/R) injury induced by tourniquet (TQ) application leads to the release of both oxygen free radicals and inflammatory cytokines. The skeletal muscle I/R may contribute to local skeletal muscle and remote organ damage affecting outcomes after total knee arthroplasty (TKA). The aim of the study is to summarize the current findings associated with I/R injury following TKA using a thigh TQ, which include cellular alterations and protective therapeutic interventions. The PubMed database was searched using the keywords "ischemia reperfusion injury," "oxidative stress," "tourniquet," and "knee arthroplasty." The search was limited to research articles published in the English language. Twenty-eight clinical studies were included in this qualitative review. Skeletal muscle I/R reduces protein synthesis, increases protein degradation, and upregulates genes in cell stress pathways. The I/R of the lower extremity elevates local and systemic oxidative stress as well as inflammatory reactions and impairs renal function. Propofol reduces oxidative injury in this I/R model. Ischemic preconditioning (IPC) and vitamin C may prevent oxygen free radical production. However, a high dose of N-acetylcysteine possibly induces kidney injury. In summary, TQ-related I/R during TKA leads to muscle protein metabolism alteration, endothelial dysfunction, oxidative stress, inflammatory response, and renal function disturbance. Propofol, IPC, and vitamin C show protective effects on oxidative and inflammatory markers. However, a relationship between biochemical parameters and postoperative clinical outcomes has not been validated.


Subject(s)
Arthroplasty, Replacement, Knee/adverse effects , Oxidative Stress , Reperfusion Injury/physiopathology , Tourniquets/adverse effects , Animals , Biomarkers , Humans , Reperfusion Injury/etiology
16.
J Cell Physiol ; 234(1): 816-824, 2018 01.
Article in English | MEDLINE | ID: mdl-30078226

ABSTRACT

Currently, electrical stimulation (ES) is used to induce changes in various tissues and cellular processes, but its effects on mitochondrial dynamics and mechanisms are unknown. The aim of this study was to compare the effects of monophasic and biphasic, anodal, and cathodal ES on apoptosis, proliferation, and mitochondrial dynamics in neuroblastoma SH-SY5Y cells. Cells were cultured and treated with ES. Alamar blue assay was performed to measure cell proliferation. The proteins expression of apoptotic-related proteins Bcl-2 associated X (Bax), B cell lymphoma 2 (Bcl-2), optic-atrophy-1 (OPA1), mitofusin2 (Mfn2), phosphorylated dynamin-related protein 1 at serine 616 (p-DRP1), and total dynamin-related protein 1 (Total-DRP1) were also determined. The results showed that monophasic anodal and biphasic anodal/cathodal (Bi Anod) ES for 1 hr at 125 pulses per minute (2.0 Hz) produced the most significant increase in cell proliferation. In addition, monophasic anodal and Bi Anod ES treated cells displayed a significant increase in the levels of anti-apoptotic protein Bcl-2, whereas the Bax levels were not changed. Moreover, the levels of Mfn2 were increased in the cells treated by Bi Anod, and OPA1 was increased by monophasic anodal and Bi Anod ES, indicating increased mitochondrial fusion in these ES-treated cells. However, the levels of mitochondrial fission indicated by DRP1 remained unchanged compared with non-stimulated cells. These findings were confirmed through visualization of mitochondria using Mitotracker Deep Red, demonstrating that monophasic anodal and Bi Anod ES could induce pro-survival effects in SH-SY5Y cells through increasing cell proliferation and mitochondrial fusion. Future research is needed to validate these findings for the clinical application of monophasic anodal and Bi Anod ES.


Subject(s)
Apoptosis/radiation effects , Cell Proliferation/radiation effects , Electric Stimulation , Mitochondrial Dynamics/radiation effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/drug effects , Dynamins , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondria/radiation effects , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Phosphorylation/genetics , Phosphorylation/radiation effects , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2-Associated X Protein/genetics
17.
Nutrition ; 54: 40-47, 2018 10.
Article in English | MEDLINE | ID: mdl-29705500

ABSTRACT

OBJECTIVES: The beneficial effects of pro-, pre-, and synbiotics on obesity with insulin resistance have been reported previously. However, the strain-specific effect of probiotics and the combination with various types of prebiotic fiber yield controversial outcomes and limit clinical applications. Our previous study demonstrated that the probiotic Lactobacillus paracasei (L. paracasei) HII01, prebiotic xylooligosaccharide (XOS), and synbiotics share similar efficacy in attenuating cardiac mitochondrial dysfunction in obese-insulin resistant rats. Nonetheless, the roles of HII01 and XOS on gut dysbiosis and gut inflammation under obese-insulin resistant conditions have not yet, to our knowledge, been investigated. Our hypothesis was that pro-, pre-, and synbiotics improve the metabolic parameters in obese-insulin resistant rats by reducing gut dysbiosis and gut inflammation. METHODS: Male Wistar rats were fed with either a normal or high-fat diet that contained 19.77% and 59.28% energy from fat, respectively, for 12 wk. Then, the high-fat diet rats were fed daily with a 108 colony forming unit of the probiotic HII01, 10% prebiotic XOS, and synbiotics for 12 wk. The metabolic parameters, serum lipopolysaccharide levels, fecal Firmicutes/Bacteroidetes ratios, levels of Enterobacteriaceae, Bifidobacteria, and gut proinflammatory cytokine gene expression were quantified. RESULTS: The consumption of probiotic L. paracasei HII01, prebiotic XOS, and synbiotics for 12 wk led to a decrease in metabolic endotoxemia, gut dysbiosis (a reduction in the Firmicutes/Bacteroidetes ratio and Enterobacteriaceae), and gut inflammation in obese-insulin resistant rats. CONCLUSIONS: Pro-, pre-, and synbiotics reduced gut dysbiosis and gut inflammation, which lead to improvements in metabolic dysfunction in obese-insulin resistant rats.


Subject(s)
Dietary Supplements , Glucuronates/administration & dosage , Lacticaseibacillus paracasei , Obesity/microbiology , Oligosaccharides/administration & dosage , Synbiotics/administration & dosage , Animals , Bifidobacterium/isolation & purification , Diet, High-Fat , Dysbiosis/microbiology , Endotoxemia/microbiology , Gastrointestinal Microbiome , Insulin Resistance , Male , Prebiotics/administration & dosage , Probiotics/administration & dosage , Rats , Rats, Wistar
18.
Free Radic Res ; 52(4): 415-425, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29424256

ABSTRACT

Trigeminal neuralgia (TN) is the neuropathic pain. Mitochondrial dysfunction, increased oxidative stress, and inflammation demonstrated in chronic pain. Carbamazepine (CBZ) is the first-line drug for TN, however, it is still insufficient. Coenzyme Q10 (CoQ10) has been used as the additional supplement for pain therapy. Nonetheless, mitochondrial respiratory proteins, oxidative stress, and inflammation in TN, and the add-on effects of CoQ10 on those defects have never been investigated. CBZ-treated TN-patients, naïve TN-patients, and control subjects were included. CBZ-treated TN-patients were randomised into two subgroups, received either CoQ10 or placebo for 2 months. Pain levels were evaluated, and peripheral blood mononuclear cells were isolated to determine the oxidative stress, mitochondrial oxidative phosphorylation (OXPHOS), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and cytokines including TNF-α, IL-1ß and IL-18 mRNA expression. Pain scales, oxidative stress, and OXPHOS levels were greater in naïve TN-patients than control, whereas the cytokine profiles were unchanged. Although pain scales were lower in CBZ-treated TN-patients than in naïve TN-patients, oxidative stress, OXPHOS, and cytokine expression profiles were not different. PGC-1α levels found to be increased in CBZ-treated TN patients when compared with the naïve group. CoQ10 supplement in CBZ-treated TN patients reduced pain scale and oxidative stress and increased antioxidants levels when compared with placebo group. However, OXPHOS, PGC-1α, and cytokines were not different between groups. These findings suggest that increased oxidative stress could be potentially involved in the pathogenesis of TN. CoQ10 supplements can reduce oxidative stress, leading to more effective pain reduction in TN patients being treated with CBZ.


Subject(s)
Mitochondrial Proteins/metabolism , Pain/drug therapy , Trigeminal Neuralgia/drug therapy , Ubiquinone/analogs & derivatives , Carbamazepine/pharmacology , Cytokines/biosynthesis , Female , Humans , Male , Middle Aged , Mitochondria/drug effects , Oxidative Stress/drug effects , Pain Management , Phosphorylation , Trigeminal Neuralgia/metabolism , Ubiquinone/pharmacology
19.
Complement Ther Clin Pract ; 23: 59-63, 2016 May.
Article in English | MEDLINE | ID: mdl-27157960

ABSTRACT

Tai Chi is a callisthenic exercise form that incorporates aerobic exercise with diaphragmatic breathing. These two aspects alone have been shown to enhance the heart rate variability, warranting research into the effects of Tai Chi on autonomic nervous system modulation and heart rate variability. A low heart rate variability has been shown to be indicative of compromised health. Any methods to enhance the heart rate variability, in particular, non-pharmacological methods, are therefore seen as beneficial to health and are sought after. The aim of this review was to comprehensively summarize the currently published studies regarding the effects of Tai Chi on heart rate variability. Both consistent and inconsistent findings are presented and discussed, and an overall conclusion attained which could benefit future clinical studies.


Subject(s)
Heart Rate/physiology , Tai Ji , Autonomic Nervous System/physiology , Humans
20.
Exp Physiol ; 101(4): 521-39, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26824522

ABSTRACT

NEW FINDINGS: What is the central question of this study? Head-to-head comparison of the therapeutic efficacy among commercial iron chelators and a dual T- (TTCC) and L-type calcium channel (LTCC) blocker on cardiac function, mitochondrial function and the protein expression of cardiac iron transporters in thalassaemic mice in iron-overloaded conditions has not been assessed. What is the main finding and its importance? The dual TTCC and LTCC blocker efonidipine could provide broad beneficial effects in the heart, liver, plasma and mitochondria in both wild-type and thalassaemic mice in iron-overloaded conditions. Its beneficial effects are of the same degree as the three commercial iron chelators currently used clinically. It is possible that efonidipine could be an alternative choice in patients unable to take iron chelators for the treatment of iron-overload conditions. Iron chelation therapy is a standard treatment in thalassaemia patients; however, its poor cardioprotective efficacy and serious side-effects are a cause for concern. Previous studies have shown that treatment with L-type calcium channel (LTCC) blockers or dual T-type calcium channel (TTCC) and LTCC blockers decreases cardiac iron and improves cardiac dysfunction in an iron-overloaded rodent model. Currently, the head-to-head comparison of therapeutic efficacy among commercial iron chelators, a dual TTCC and LTCC blocker and an LTCC blocker on cardiac function, mitochondrial function and the protein expression of cardiac iron transporters in thalassaemic mice in an iron-overloaded state has never been investigated. An iron-overloaded state was induced in ß-thalassaemic and wild-type mice. Cardiac iron overload was induced to a greater extent than in a previous study by feeding the mice with an iron-enriched diet for 4 months. Then, an LTCC blocker (amlodipine) or a dual TTCC and LTCC blocker (efonidipine) or one of the commercial iron chelators (deferoxamine, deferasirox or deferiprone) was administered for 1 month with continuous iron feeding. All treatments reduced cardiac iron deposition and improved mitochondrial and cardiac dysfunction in both types of mice. Only efonidipine and the iron chelators reduced liver iron accumulation, liver malondialdehyde and plasma malondialdehyde in these mice. Although all pharmacological interventions reduced cardiac iron deposition, they did not alter the protein expression levels of cardiac iron transporter. These findings indicated that efonidipine provided all benefits to the same degree as the three commercial iron chelators. These findings indicate that a dual TTCC and LTCC blocker could be beneficial for treatment of an iron-overloaded state.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Cardiovascular Diseases/drug therapy , Heart/drug effects , Iron Overload/drug therapy , Thalassemia/drug therapy , Animals , Benzoates/pharmacology , Cardiovascular Diseases/metabolism , Deferasirox , Deferiprone , Deferoxamine/pharmacology , Dihydropyridines/pharmacology , Iron Chelating Agents/pharmacology , Iron Overload/metabolism , Male , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Nitrophenols/pharmacology , Organophosphorus Compounds/pharmacology , Pyridones/pharmacology , Thalassemia/metabolism , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL