Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Dig Dis Sci ; 69(4): 1293-1301, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38363519

ABSTRACT

BACKGROUND: The relationship between Helicobacter pylori (H. pylori) infection and small intestinal bacterial overgrowth (SIBO) has attracted attention recently. AIMS: To analyze the influence of H. pylori infection and eradication on SIBO, IMO, and abdominal symptoms. METHODS: Patients with gastrointestinal symptoms were tested for 13C urea breath test and if positive, treated with bismuth-based quadruple therapy. Lactulose hydrogen methane breath test (HMBT) was performed and symptoms were assessed using gastrointestinal symptom rating scale (GSRS) before and 6 weeks after eradication. RESULTS: Of the 102 subjects, 53 were H. pylori positive. The prevalence of SIBO and IMO were higher in patients with H. pylori infection than in those without infection (49.1% vs 24.5%, P = 0.019 for SIBO; 24.5% vs 8.2%, P = 0.027 for IMO). GSRS scores were similar between H. pylori-infected and uninfected patients (2 (IQR: 1;3) vs 2 (IQR: 1;2), P = 0.211). Patients with SIBO or IMO presented higher GSRS scores than patients with both SIBO and IMO negative (2 (IQR: 2;3), 2 (IQR: 2;3) vs 2 (IQR: 1;2), P = 0.011, 0.001, respectively). For the 50 patients who successfully eradicated H. pylori, the response rates for SIBO and IMO were 66.7% and 76.9%, respectively. GSRS scores also significantly decreased (2 (IQR: 1;3) to 0 (IQR: 0;1), P < 0.001) after eradication. CONCLUSION: Helicobacter pylori infection was associated with higher prevalence of SIBO and IMO, both of which led to more pronounced abdominal symptoms. H. pylori eradication also achieved therapeutic effects on SIBO and IMO, accompanied by relief of abdominal symptoms.


Subject(s)
Gastrointestinal Diseases , Helicobacter Infections , Helicobacter pylori , Humans , Helicobacter Infections/drug therapy , Gastrointestinal Diseases/microbiology , Bismuth/therapeutic use , Lactulose/therapeutic use , Breath Tests , Anti-Bacterial Agents/therapeutic use , Drug Therapy, Combination
2.
J Cardiovasc Pharmacol ; 70(2): 102-109, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28437280

ABSTRACT

Chronic intermittent hypoxia (CIH), the hallmark of obstructive sleep apnea syndrome (OSAS), has been reported to play a key role in the development of OSAS-associated cardiovascular diseases including cardiac remodeling. RhoA/Rho-kinase (ROCK) pathway has also been implicated in myocardial remodeling, but the exact mechanisms are not fully elucidated. This study's purpose is to investigate the influence of fasudil, a selective ROCK inhibitor, on CIH-induced left ventricular remodeling in rats and its possible mechanisms. Adult male Sprague-Dawley rats suffered from CIH or normoxia stimulus and were intervened with vehicle or fasudil (10 mg·kg·d, intraperitoneal injection) for 6 weeks. In this study, treatment with fasudil significantly reversed intermittent hypoxia-induced histopathological transformations and ultrastructural changes in rat myocardium. Moreover, fasudil downregulated the protein levels of RhoA and phosphorylation of myosin phosphatase targeting subunit-1 (MYPT1), thus effectively inhibited the activation of RhoA/ROCK signaling pathway. Simultaneously, activity of nuclear factor (NF)-kB was suppressed by fasudil, which was accompanied by reduced NF-kB downstream inflammatory genes including interleukin-6, tumor necrosis factor-a and monocyte chemotactic protein-1, and apoptosis. These results suggest that fasudil attenuates myocardial remodeling in CIH rats, at least partly by suppressing activation of NF-kB. Inhibition of the RhoA/ROCK pathway could become an important therapeutic target in the prevention of OSAS-related cardiomyopathy.


Subject(s)
Apoptosis/physiology , Hypoxia/metabolism , Myocardium/metabolism , Protein Kinase Inhibitors/pharmacology , Ventricular Remodeling/physiology , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Apoptosis/drug effects , Hypoxia/drug therapy , Hypoxia/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Myocardium/pathology , Protein Kinase Inhibitors/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology , Ventricular Remodeling/drug effects , rho-Associated Kinases/antagonists & inhibitors
3.
Arch Biochem Biophys ; 606: 41-52, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27412517

ABSTRACT

Autophagy is tightly regulated to maintain cardiac homeostasis. Impaired autophagy is closely associated with pathological cardiac hypertrophy. However, the relationship between autophagy and cardiac hypertrophy induced by chronic intermittent hypoxia (CIH) is not known. In the present study, we measured autophagy-related genes and autophagosomes during 10 weeks of CIH in rats, and 6 days in H9C2 cardiomyocytes, and showed that autophagy was impaired. This conclusion was confirmed by the autophagy flux assay. We detected significant hypertrophic changes in myocardium with impaired autophagy. Rapamycin, an autophagy enhancer, attenuated the cardiac hypertrophy induced by CIH. Moreover, silencing autophagy-related gene 5 (ATG5) exerted the opposite effect. The role of adenosine monophosphate-activated protein kinase (AMPK) in regulating autophagy under CIH was confirmed using AICAR to upregulate this enzyme and restore autophagy flux. Restoring autophagy by AICAR or rapamycin significantly reversed the hypertrophic changes in cardiomyocytes. To investigate the mechanism of autophagy impairment, we compared phospho (p)-AMPK, p-Akt, cathepsin D, and NFAT3 levels, along with calcineurin activity, between sham and CIH groups. CIH activated calcineurin, and inhibited AMPK and AMPK-mediated autophagy in an Akt- and NFAT3-independent manner. Collectively, these data demonstrated that impaired autophagy induced by CIH through the AMPK pathway contributed to cardiac hypertrophy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Cardiomegaly/pathology , Hypoxia , Adenosine Triphosphate/chemistry , Adenylate Kinase/metabolism , Animals , Apoptosis , Autophagy-Related Protein 5/metabolism , Calcineurin/metabolism , Cardiomegaly/metabolism , Hemodynamics , Male , Microscopy, Electron, Transmission , Myocardium/metabolism , Phosphorylation , RNA Interference , Rats , Rats, Sprague-Dawley , Sirolimus/chemistry
4.
Chin Med J (Engl) ; 128(16): 2168-75, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26265609

ABSTRACT

BACKGROUND: The mechanism of the neural injury caused by chronic intermittent hypoxia (CIH) that characterizes obstructive sleep apnea syndrome (OSAS) is not clearly known. The purpose of this study was to investigate whether P2X7 receptor (P2X7R) is responsible for the CIH-induced neural injury and the possible pathway it involves. METHODS: Eight-week-old male C57BL/6 mice were used. For each exposure time point, eight mice divided in room air (RA) and IH group were assigned to the study of P2X7R expression. Whereas in the 21 days-Brilliant Blue G (BBG, a selective P2X7R antagonist) study, 48 mice were randomly divided into CIH group, BBG-treated CIH group, RA group and BBG-treated RA group. The hippocampus P2X7R expression was determined by Western blotting and real-time polymerase chain reaction (PCR). The spatial learning was analyzed by Morris water maze. The nuclear factor kappa B (NFκB) and NADPH oxidase 2 (NOX2) expressions were analyzed by Western blotting. The expressions of tumor necrosis factor α, interleukin 1ß (IL-ß), IL-18, and IL-6 were measured by real-time PCR. The malondialdehyde and superoxide dismutase levels were detected by colorimetric method. Cell damage was evaluated by Hematoxylin and Eosin staining and Terminal Transferase dUTP Nick-end Labeling method. RESULTS: The P2X7R mRNA was elevated and sustained after 3-day IH exposure and the P2X7R protein was elevated and sustained after 7-day IH exposure. In the BBG study, the CIH mice showed severer neuronal cell damage and poorer performance in the behavior test. The increased NFκB and NOX2 expressions along with the inflammation injury and oxidative stress were also observed in the CIH group. BBG alleviated CIH-induced neural injury and consequent functional deficits. CONCLUSIONS: The P2X7R antagonism attenuates the CIH-induced neuroinflammation, oxidative stress, and spatial deficits, demonstrating that the P2X7R is an important therapeutic target in the cognition deficits accompanied OSAS.


Subject(s)
Receptors, Purinergic P2X7/physiology , Sleep Apnea, Obstructive/metabolism , Animals , Disease Models, Animal , Hypoxia , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/analysis , Rosaniline Dyes/pharmacology
5.
Urology ; 86(4): 844.e7-844.e14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26206453

ABSTRACT

OBJECTIVE: To conduct a study using a rodent model of chronic intermittent hypoxia (CIH) to define whether endoplasmic reticulum stress (ERS) is involved in the CIH-induced apoptosis of penile tissue and erectile dysfunction (ED), and whether treatment with N-acetylcysteine (NAC) alleviates pathological variations in corpus cavernosa. Previous work has prompted that CIH acted as the major trigger linking obstructive sleep apnea syndrome and ED. MATERIALS AND METHODS: Five-month-old Sprague-Dawley male rats were subjected to 8 hours of intermittent hypoxia per day, with or without NAC for 5 weeks. Erectile function, apoptosis of penile tissue, levels of ERS-associated proapoptotic effectors, and nitric oxide (NO) and nitric oxide synthase (NOS) activity were determined. RESULTS: Treatment with NAC inhibited apoptosis of penile tissue, the expressions of ERS-related products: BIP, CHOP, caspase12, and Bax, NO, and endothelial NOS. Administration of NAC before CIH significantly improved the CIH-induced impaired erectile function. CONCLUSION: Our results show that pre-CIH NAC administration ameliorates the ED following CIH partly by alleviating CIH-induced ERS and cell apoptosis via regulating the expressions of BIP, CHOP, caspase12, and Bax.


Subject(s)
Acetylcysteine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Erectile Dysfunction/drug therapy , Hypoxia/complications , Animals , Apoptosis , Blotting, Western , Disease Models, Animal , Erectile Dysfunction/etiology , Erectile Dysfunction/metabolism , Free Radical Scavengers/pharmacology , Hypoxia/pathology , Immunohistochemistry , Male , Nitric Oxide Synthase/metabolism , Rats , Rats, Sprague-Dawley
6.
Respir Physiol Neurobiol ; 218: 57-63, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26200444

ABSTRACT

Hippocampal neuronal damage is critical for the initiation and progression of neurocognitive impairment accompanied obstructive sleep apnea syndrome (OSAS). Toll-like receptor 4 (TLR4) plays an important role in the development of several hippocampus-related neural disorders. Atorvastatin was reported beneficially regulates TLR4. Here, we examined the effects of atorvastatin on hippocampal injury caused by chronic intermittent hypoxia (CIH), the most characteristic pathophysiological change of OSAS. Mice were exposed to intermittent hypoxia with or without atorvastatin for 4 weeks. Cell damage, the expressions of TLR4 and its two downstream factors myeloid differentiation factor 88 (MYD88) and TIR-domain-containing adapter-inducing interferon-ß (TRIF), inflammatory agents (tumor necrosis factor α and interleukin 1ß), and the oxidative stress (superoxide dismutase and malondialdehyde) were determined. Atorvastatin decreased the neural injury and the elevation of TLR4, MyD88, TRIF, pro-inflammatory cytokines and oxidative stress caused by CIH. Our study suggests that atorvastatin may attenuate CIH induced hippocampal neuronal damage partially via TLR4 and its downstream signaling pathway.


Subject(s)
Atorvastatin/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sleep Apnea, Obstructive/drug therapy , Toll-Like Receptor 4/metabolism , Animals , Chronic Disease , Disease Models, Animal , Hippocampus/pathology , Hippocampus/physiopathology , Hypoxia/drug therapy , Hypoxia/pathology , Hypoxia/physiopathology , Interleukin-1beta/metabolism , Male , Malondialdehyde/metabolism , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Neurons/pathology , Neurons/physiology , Random Allocation , Signal Transduction/drug effects , Sleep Apnea, Obstructive/pathology , Sleep Apnea, Obstructive/physiopathology , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Int J Mol Med ; 36(1): 150-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25936416

ABSTRACT

Obstructive sleep apnea, characterized by recurrent episodes of hypoxia [intermittent hypoxia (IH)], has been identified as a risk factor for cardiovascular diseases. The O-linked ß-N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation) of proteins has important regulatory implications on the pathophysiology of cardiovascular disorders. In this study, we examined the role of O-GlcNAcylation in cardiac architecture and left ventricular function following IH. Rats were randomly assigned to a normoxia and IH group (2 min 21% O2; 2 min 6-8% O2). Left ventricular function, myocardial morphology and the levels of signaling molecules were then measured. IH induced a significant increase in blood pressure, associated with a gradually abnormal myocardial architecture. The rats exposed to 2 or 3 weeks of IH presented with augmented left ventricular systolic and diastolic function, which declined at week 4. Consistently, the O-GlcNAc protein and O-GlcNAcase (OGA) levels in the left ventricular tissues steadily increased following IH, reaching peak levels at week 3. The O-GlcNAc transferase (OGT), extracellular signal-regulated kinase 1/2 (ERK1/2) and the p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation levels were affected in an opposite manner. The phosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII) remained unaltered. In parallel, compared with exposure to normoxia, 4 weeks of IH augmented the O-GlcNAc protein, OGT, phosphorylated ERK1/2 and p38 MAPK levels, accompanied by a decrease in OGA levels and an increase in the levels of myocardial nuclear factor-κB (NF-κB), inflammatory cytokines, caspase-3 and cardiomyocyte apoptosis. Taken together, our suggest a possible involvement of O-GlcNAc protein and MAPK signaling in the alterations of left ventricular function and cardiac injury following IH.


Subject(s)
Acetylglucosamine/metabolism , Cardiovascular Diseases/physiopathology , Cell Hypoxia/physiology , MAP Kinase Signaling System/physiology , Ventricular Function, Left/physiology , Animals , Apoptosis/physiology , Blood Gas Analysis , Blood Pressure/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caspase 3/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , In Situ Nick-End Labeling , Interleukin-6/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , N-Acetylglucosaminyltransferases/metabolism , NF-kappa B/metabolism , Phosphorylation , Rats , Rats, Wistar , Sleep Apnea, Obstructive/physiopathology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Brain Res ; 1596: 48-57, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25463026

ABSTRACT

Obstructive sleep apnea syndrome (OSAS) plays a critical role in the initiation and progression of Alzheimer׳s disease (AD), but little is known about the precise mechanism of OSAS-induced AD. Nitric oxide synthase (NOS) and nitric oxide (NO) are known to play key roles in the development of AD. Several studies have confirmed that an angiotensin II type 1 receptor blocker, telmisartan, beneficially regulates NOS and NO. Here, we examined the neuroprotective effects of telmisartan against hippocampal apoptosis induced by chronic intermittent hypoxia (CIH), the most characteristic pathophysiological change of OSAS. Adult male Sprague Dawley rats were subjected to 8h of intermittent hypoxia per day with or without telmisartan for eight weeks. Neuronal apoptosis in the hippocampal CA1 region, NOS activity, NO content, and the presence of inflammatory agents and radical oxygen species in the hippocampus were determined. The results showed that CIH activated inducible nitric oxide synthase (iNOS), increased NO content, and enhanced lipid peroxidation and inflammatory responses in the hippocampus. Treatment with telmisartan inhibited excessive iNOS and NO generation and reduced lipid peroxidation and inflammatory responses. In addition, telmisartan significantly ameliorated the hippocampal apoptosis induced by CIH. In conclusion, Pre-CIH telmisartan administration attenuated CIH-induced hippocampal apoptosis partly by regulating NOS activity, inhibiting excessive NO generation, and reducing lipid peroxidation and inflammatory responses.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Benzoates/pharmacology , Hippocampus/pathology , Hypoxia/complications , Hypoxia/pathology , Inflammation/etiology , Lipid Peroxidation/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Analysis of Variance , Animals , Apoptosis/physiology , C-Reactive Protein/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , In Situ Nick-End Labeling , Interleukin-6/blood , Leukocyte Common Antigens/metabolism , Lipid Peroxidation/physiology , Male , Rats , Rats, Sprague-Dawley , Telmisartan
9.
Sleep Breath ; 19(2): 703-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25413957

ABSTRACT

PURPOSE: NO and NO synthase (NOS) are known to play key roles in the development of myocardial apoptosis induced by ischemia/hypoxia. Current evidence suggests that angiotensin II type 1 receptor blockers, such as telmisartan, lower blood pressure and produce beneficial regulatory effects on NO and NOS. Here, we examined the protective role of telmisartan in myocardial apoptosis induced by chronic intermittent hypoxia (CIH). METHODS: Adult male Sprague-Dawley rats were subjected to 8 h of intermittent hypoxia/day, with/without telmisartan for 8 weeks. Myocardial apoptosis, NO and NOS activity, and levels of inflammatory mediators and radical oxygen species were determined. RESULTS: Treatment with telmisartan preserved endothelial NOS expression and inhibited inducible NOS and excessive NO generation, while reducing oxidation/nitration stress and inflammatory responses. Administration of telmisartan before CIH significantly ameliorated the CIH-induced myocardial apoptosis. CONCLUSIONS: This study show that pre-CIH telmisartan administration ameliorated myocardial injury following CIH by attenuating CIH-induced myocardial apoptosis via regulation of NOS activity and inhibition of excessive NO generation, oxidation/nitration stress, and inflammatory responses.


Subject(s)
Apoptosis/drug effects , Benzimidazoles/pharmacology , Benzoates/pharmacology , Hypoxia/pathology , Inflammation Mediators/metabolism , Myocardium/cytology , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Sleep Apnea, Obstructive/pathology , Animals , C-Reactive Protein/metabolism , Disease Models, Animal , Interleukin-6/blood , Lipid Peroxidation/drug effects , Male , Nitric Oxide Synthase/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Telmisartan
10.
Biochem Biophys Res Commun ; 446(1): 292-7, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24582748

ABSTRACT

Inflammatory processes and oxidative stress are known to play a key role in the development of cardiovascular complications such as cardiac hypertrophy induced by chronic intermittent hypoxia (CIH), the most characteristic pathophysiological change of obstructive sleep apnea syndrome (OSAS). Current evidence suggests that competitive inhibitors of 3-hydroxy-3-methylglutaryl-CoA coenzyme A reductase, such as atorvastatin, not only reduce blood lipids but also have anti-inflammatory and inhibit oxidative stress benefits. This study examined the protective role of atorvastatin in CIH-induced cardiac hypertrophy. Adult male wistar rats were subjected to 8h of intermittent hypoxia/day, with/without atorvastatin for 6 weeks. Ventricular remodeling, toll-like receptor 4 (TLR-4), myeloid differentiation primary response protein 88 (MYD88), inflammatory agents and radical oxygen species were determined. As a result, we found that treatment with atorvastatin markedly inhibited the mRNA and protein expressions of TLR4, MYD88 and the downstream inflammatory agents and radical oxygen species. Administration of atorvastatin following CIH significantly ameliorated the myocardial injury, such as cardiac hypertrophy. In conclusion, Pre-CIH atorvastatin administration may attenuate TLR-4/MYD88 mediated inflammatory processes and oxidative stress in the injured rat myocardium, and this may be one mechanism by which atorvastatin ameliorated myocardial injury following CIH.


Subject(s)
Heptanoic Acids/pharmacology , Hypoxia/physiopathology , Myeloid Differentiation Factor 88/metabolism , Myocardium/metabolism , Myocardium/pathology , Pyrroles/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Atorvastatin , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Cytokines/genetics , Heart/drug effects , Hypoxia/complications , Male , Myeloid Differentiation Factor 88/genetics , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sleep Apnea, Obstructive/congenital , Sleep Apnea, Obstructive/physiopathology , Toll-Like Receptor 4/genetics , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...