Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
JAAD Int ; 10: 89-94, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36691658

ABSTRACT

Background: Hidradenitis suppurativa (HS) is a chronic debilitating inflammatory skin disorder known to result in significant psychological symptoms and impaired quality of life. However, most of these studies are limited to western countries, with limited data from other sociocultural regions. Objective: To understand the psychosocial burden of HS in the Asian context, by exploring the correlation between objective disease measures with psychosocial health and work productivity. Methods: A prospective single-center questionnaire study was conducted. A total of 45 patients with HS completed a questionnaire and examination by a dermatologist. Results: Higher objective disease severity scores (Hurley, physician global assessment, International Hidradenitis Suppurative Severity, and modified Sartorius) correlated with poorer quality of life (Dermatology Life Quality Index and Patient Global Assessment), increased anxiety and depression (hospital anxiety and depression scale) and at-work productivity loss. There was no significant correlation between objective disease severity and other domains Work productivity and activity impairment score or self-esteem (Rosenburg Self-Esteem Scale). Limitations: The main limitation of our study is the small sample size, assessment at a single time point, and lack of control cohort. Conclusion: Our findings demonstrate the impact of HS on a patient's psychosocial well-being and emphasize the importance of treating patients from a holistic standpoint.

2.
Exp Ther Med ; 25(1): 27, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36561615

ABSTRACT

Proteinuria is a common clinical manifestation of kidney diseases, such as glomerulonephritis, nephrotic syndrome, immunoglobulin A nephropathy and diabetic nephropathy. Therefore, proteinuria is considered to be a risk factor for renal dysfunction. Furthermore, proteinuria is also significantly associated with the progression of kidney diseases and increased mortality. Its occurrence is closely associated with damage to the structure of the glomerular filtration membrane. An impaired glomerular filtration membrane can affect the selective filtration function of the kidneys; therefore, several macromolecular substances, such as proteins, may pass through the filtration membrane and promote the manifestation of proteinuria. It has been reported that ion channels play a significant role in the mechanisms underlying proteinuria. Ion channel mutations or other dysfunctions have been implicated in several diseases, therefore ion channels could be used as major therapeutic targets. The mechanisms underlying the action of ion channels and ion transporters in proteinuria have been overlooked in the literature, despite their importance in identifying novel targets for treating proteinuria and delaying the progression of kidney diseases. The current review article focused on the four key ion channel groups, namely Na+, Ca2+, Cl- and K+ ion channels and the associated ion transporters.

3.
Inflammation ; 46(2): 556-572, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36269513

ABSTRACT

Neuropsychiatric systemic lupus erythematosus (NPSLE) is a serious complication of systemic lupus erythematosus (SLE) involving the nervous system with high morbidity and mortality. A key hypothesis in NPSLE is that a disrupted barrier allows autoantibodies and immune components of peripheral blood to penetrate into the central nervous system (CNS), resulting in inflammation and damage. The blood cerebrospinal fluid barrier (BCSFB), which consists of the choroid plexus and the hypothalamic tanycytes, has long been regarded as an immunological sanctuary site. 1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] is the active form of vitamin D, which plays multiple roles in inflammation and immunoregulation. In this study, we investigated the possible protective effects of 1,25-dihydroxyvitamin D3 against BCSFB dysfunction in NPSLE in MRL/lpr mice and explored the mechanism by which 1,25-dihydroxyvitamin D3 inhibits the progression of NPSLE. In this study, we found that supplementation with 1,25-dihydroxyvitamin D3 markedly improved serological and immunological indices, delayed inflammatory infiltration, delayed neuronal deformation, and upregulated the expression of brain-derived neurotrophic factor (BDNF) proteins in the brain. Furthermore, 1,25-dihydroxyvitamin D3 downregulated proinflammatory cytokines such as nuclear factor kappa-B (NF-κB) and tumor necrosis factor-α (TNF-α) by activating peroxisome proliferator-activated receptor γ (PPARγ), and it reduced the expression of the TGF-ß/Smad signaling pathway. Our findings demonstrate that 1,25-dihydroxyvitamin D3 delayed cell infiltration into the choroid plexus and decreased markers suggestive of cognitive decline in MRL/lpr mice, and the mechanism may be related to protection against BCSFB disruption through activation of the anti-inflammatory PPARγ/NF-κB/TNF-α pathway as well as upregulation of BDNF and inhibition of the TGF-ß/Smad signaling pathway. These findings provide a novel direction for the study of NPSLE.


Subject(s)
Lupus Erythematosus, Systemic , Tumor Necrosis Factor-alpha , Animals , Mice , Brain-Derived Neurotrophic Factor , Calcitriol/pharmacology , Calcitriol/therapeutic use , Choroid Plexus , Inflammation/drug therapy , Inflammation/complications , Lupus Erythematosus, Systemic/therapy , Mice, Inbred MRL lpr , NF-kappa B , PPAR gamma , Transforming Growth Factor beta , Smad Proteins/metabolism
4.
Redox Biol ; 51: 102266, 2022 05.
Article in English | MEDLINE | ID: mdl-35217293

ABSTRACT

BACKGROUND: Renal fibrosis, associated with oxidative stress and nitric oxide (NO) deficiency, contributes to the development of chronic kidney disease and renal failure. As major energy source in maintaining renal physiological functions, tubular epithelial cells with decreased fatty acid oxidation play a key role in renal fibrosis development. Inorganic nitrate, found in high levels in certain vegetables, can increase the formation and signaling by bioactive nitrogen species, including NO, and dampen oxidative stress. In this study, we evaluated the therapeutic value of inorganic nitrate treatment on development of kidney fibrosis and investigated underlying mechanisms including regulation of lipid metabolism in tubular epithelial cells. METHODS: Inorganic nitrate was supplemented in a mouse model of complete unilateral ureteral obstruction (UUO)-induced fibrosis. Inorganic nitrite was applied in transforming growth factor ß-induced pro-fibrotic cells in vitro. Metformin was administrated as a positive control. Fibrosis, oxidative stress and lipid metabolism were evaluated. RESULTS: Nitrate treatment boosted the nitrate-nitrite-NO pathway, which ameliorated UUO-induced renal dysfunction and fibrosis in mice, represented by improved glomerular filtration and morphological structure and decreased renal collagen deposition, pro-fibrotic marker expression, and inflammation. In human proximal tubule epithelial cells (HK-2), inorganic nitrite treatment prevented transforming growth factor ß-induced pro-fibrotic changes. Mechanistically, boosting the nitrate-nitrite-NO pathway promoted AMP-activated protein kinase (AMPK) phosphorylation, improved AKT-mediated peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) activity and restored mitochondrial function. Accordingly, treatment with nitrate (in vivo) or nitrite (in vitro) decreased lipid accumulation, which was associated with dampened NADPH oxidase activity and mitochondria-derived oxidative stress. CONCLUSIONS: Our findings indicate that inorganic nitrate and nitrite treatment attenuates the development of kidney fibrosis by targeting oxidative stress and lipid metabolism. Underlying mechanisms include modulation of AMPK and AKT-PGC1α pathways.


Subject(s)
Kidney Diseases , Ureteral Obstruction , AMP-Activated Protein Kinases/metabolism , Animals , Female , Fibrosis , Humans , Kidney/metabolism , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Kidney Diseases/metabolism , Lipid Metabolism , Male , Mice , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/metabolism
5.
Front Endocrinol (Lausanne) ; 12: 740200, 2021.
Article in English | MEDLINE | ID: mdl-34956075

ABSTRACT

The role of obesity in the development of dorsopathies is still unclear. In this study, we assessed the associations between body mass index (BMI) and several dorsopathies including intervertebral disc degeneration (IVDD), low back pain (LBP), and sciatica by using the Mendelian randomization method. We also assessed the effect of several obesity-related traits on the same outcomes. Single-nucleotide polymorphisms associated with the exposures are extracted from summary-level datasets of previously published genome-wide association studies. Summary-level results of IVDD, LBP, and sciatica were from FinnGen. In our univariable Mendelian randomization analysis, BMI is significantly associated with increased risks of all dorsopathies including sciatica (OR = 1.33, 95% CI, 1.21-1.47, p = 5.19 × 10-9), LBP (OR = 1.28, 95% CI, 1.18-1.39, p = 6.60 × 10-9), and IVDD (OR = 1.23, 95% CI, 1.14-1.32, p = 2.48 × 10-8). Waist circumference, hip circumference, whole-body fat mass, fat-free mass, and fat percentage, but not waist-hip ratio, were causally associated with increased risks of IVDD and sciatica. Higher hip circumference, whole-body fat mass, fat-free mass, and fat percentage increased the risk of LBP. However, only whole-body fat-free mass remained to have a significant association with the risk of IVDD after adjusting for BMI with an odds ratio of 1.57 (95% CI, 1.32-1.86, p = 2.47 × 10-7). Proportions of BMI's effect on IVDD, sciatica, and LBP mediated by leisure sedentary behavior were 41.4% (95% CI, 21.8%, 64.8%), 33.8% (95% CI, 17.5%, 53.4%), and 49.7% (95% CI, 29.4%, 73.5%), respectively. This study provides evidence that high BMI has causal associations with risks of various dorsopathies. Weight control is a good measure to prevent the development of dorsopathies, especially in the obese population.


Subject(s)
Intervertebral Disc Degeneration/complications , Low Back Pain/complications , Obesity/complications , Polymorphism, Single Nucleotide , Sciatica/complications , Adult , Body Mass Index , Humans , Intervertebral Disc Degeneration/genetics , Low Back Pain/genetics , Mendelian Randomization Analysis , Obesity/genetics , Sciatica/genetics , Waist Circumference , Waist-Hip Ratio
6.
Redox Biol ; 39: 101836, 2021 02.
Article in English | MEDLINE | ID: mdl-33360353

ABSTRACT

BACKGROUND: Renal ischemia-reperfusion (IR) injury is a common cause of acute kidney injury (AKI), which is associated with oxidative stress and reduced nitric oxide (NO) bioactivity and increased risk of developing chronic kidney disease (CKD) and cardiovascular disease (CVD). New strategies that restore redox balance may have therapeutic implications during AKI and associated complications. AIM: To investigate the therapeutic value of boosting the nitrate-nitrite-NO pathway during development of IR-induced renal and cardiovascular dysfunction. METHODS: Male C57BL/6 J mice were given sodium nitrate (10 mg/kg, i. p) or vehicle 2 h prior to warm ischemia of the left kidney (45 min) followed by sodium nitrate supplementation in the drinking water (1 mmol/kg/day) for the following 2 weeks. Blood pressure and glomerular filtration rate were measured and blood and kidneys were collected and used for biochemical and histological analyses as well as renal vessel reactivity studies. Glomerular endothelial cells exposed to hypoxia-reoxygenation, with or without angiotensin II, were used for mechanistic studies. RESULTS: IR was associated with reduced renal function and slightly elevated blood pressure, in combination with renal injuries, inflammation, endothelial dysfunction, increased Ang II levels and Ang II-mediated vasoreactivity, which were all ameliorated by nitrate. Moreover, treatment with nitrate (in vivo) and nitrite (in vitro) restored NO bioactivity and reduced mitochondrial oxidative stress and injuries. CONCLUSIONS: Acute treatment with inorganic nitrate prior to renal ischemia may serve as a novel therapeutic approach to prevent AKI and CKD and associated risk of developing cardiovascular dysfunction.


Subject(s)
Nitrates , Reperfusion Injury , Animals , Endothelial Cells , Ischemia/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Nitrates/metabolism , Oxidative Stress , Reperfusion , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
7.
Exp Ther Med ; 20(3): 2049-2054, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32782516

ABSTRACT

Expression of syndecan-1 (SDC-1) in rats with acute kidney injury and the protective effect of GM6001 on the kidney were investigated. Fifty SD rats were selected and randomly divided into control group (CG) (n=15), treatment control group (TCG) (n=10), module group (MG) (n=15) and treatment group (TG) (n=10). In TG, the model of acute renal injury (AKI) in rats was established after pretreatment of intraperitoneal injection of GM6001 one day before modeling. In MG, the same amount of saline was injected intraperitoneally one day before modeling and the same treatment was done on the day of modeling. In CG, the same amount of saline was injected intraperitoneally one day before modeling but the model was not made. In TCG, rats were pretreated with intraperitoneal injection of GM6001 one day before modeling but the model was not made. The contents of blood urea nitrogen (BUN) in serum, serum creatinine (SCR), uric acid (UA) and blood ß2-microglobulin (ß2-MG) were detected by ELISA. The content of SDC-1 in renal tissues was detected by qRT-PCR and western blotting. Expression of SDC-1 in renal tissue of 24 rats after modeling was lower than that of MG (P<0.050). SDC-1 expression was the highest in TG (P<0.05). Compared with before modeling, the contents of BUN, SCR, UA and ß2-MG in MG and TG increased (P<0.05). After modeling, the contents of serum BUN, SCR, UA and ß2-MG in TG were significantly lower than those in MG (P<0.05). The levels of SDC-1 in renal tissue of rats with acute kidney injury increased. After GM6001 treatment, SDC-1 levels can be improved and has a certain protective effect on the kidneys.

8.
Nitric Oxide ; 97: 48-56, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32032718

ABSTRACT

BACKGROUND/PURPOSE: Unhealthy dietary habits contribute to the increasing incidence of metabolic syndrome and type 2 diabetes (T2D), which is accompanied by oxidative stress, compromised nitric oxide (NO) bioavailability and increased cardiovascular risk. Apart from lifestyle changes, biguanides such as metformin are the first-line pharmacological treatment for T2D. Favourable cardiometabolic effects have been demonstrated following dietary nitrate supplementation to boost the nitrate-nitrite-NO pathway. Here we aim to compare the therapeutic value of inorganic nitrate and metformin alone and their combination in a model of cardiometabolic disease. EXPERIMENTAL APPROACH: Mice were fed control or high fat diet (HFD) for 7 weeks in combination with the NO synthase (NOS) inhibitor l-NAME to induce metabolic syndrome. Simultaneously, the mice were treated with vehicle, inorganic nitrate, metformin or a combination of nitrate and metformin in (drinking water). Cardiometabolic functions were assessed in vivo and tissues were collected/processed for analyses. KEY RESULTS: HFD + L-NAME was associated with cardiometabolic dysfunction, compared with controls, as evident from elevated blood pressure, endothelial dysfunction, impaired insulin sensitivity and compromised glucose clearance as well as liver steatosis. Both nitrate and metformin improved insulin/glucose homeostasis, whereas only nitrate had favourable effects on cardiovascular function and steatosis. Mechanistically, metformin and nitrate improved AMPK signalling, whereas only nitrate attenuated oxidative stress. Combination of nitrate and metformin reduced HbA1c and trended to further increase AMPK activation. CONCLUSION/IMPLICATIONS: Nitrate and metformin had equipotent metabolic effects, while nitrate was superior regarding protection against cardiovascular dysfunction and liver steatosis. If reproduced in future clinical trials, these findings may have implications for novel nutrition-based strategies against metabolic syndrome, T2D and associated complications.


Subject(s)
Cardiovascular Diseases/drug therapy , Disease Models, Animal , Metformin/therapeutic use , Nitrates/therapeutic use , Administration, Oral , Animals , Cardiovascular Diseases/metabolism , Diet, High-Fat/adverse effects , Enzyme Inhibitors/pharmacology , Male , Metformin/administration & dosage , Metformin/metabolism , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/administration & dosage , Nitrates/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism
9.
Int J Mol Med ; 45(3): 731-742, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31985025

ABSTRACT

Ligustrazine has been used to alleviate clinical acute kidney injury (AKI); however, the underlying molecular mechanisms are poorly understood. In order to further elucidate the molecular mechanism underlying its occurrence, the role of nucleotide­binding oligomerization domain­containing 2 (NOD2) in AKI was investigated in the present study, and the results indicated that ligustrazine exerts an important protective effect against AKI in vivo by inhibiting the upregulation of NOD2 expression and reducing apoptosis of kidney cells following ischemia/reperfusion injury in rat models. Furthermore, the inhibitory role of ligustrazine on the upregulation of NOD2 and apoptosis of kidney cells induced by CoCl2 and oxygen and glucose deprivation followed by reoxygenation was investigated in in vitro experiments. The effect of ligustrazine on NOD2 downregulation was partially blocked by inhibiting autophagy. To the best of our knowledge, the results of the present study are the first to provide evidence that ligustrazine can inhibit NOD2­mediated inflammation to protect against renal injury, which may be in part attributed to the induction of autophagy. These findings may help design and develop new approaches and therapeutic strategies for AKI to prevent the deterioration of renal function.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Pyrazines/therapeutic use , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Male , Nod2 Signaling Adaptor Protein/genetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Ann Clin Lab Sci ; 49(6): 756-762, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31882426

ABSTRACT

Hyperuricemia (HUA) is positively correlated with the progression of cardiovascular and metabolic diseases. Anti-HUA drugs aim to either reduce uric acid production or promote uric acid excretion. Urate transporter 1 (URAT1) is a major urate transporter involved in renal uric acid reabsorption and excretion, making it an important anti-HUA drug target. To better understand the characteristics of URAT1 under pathological conditions, the present study aims to investigate URAT1 modulation in HUA mouse and cell line models. We found that URAT1 expression increased in the kidneys of HUA mice with normal renal function, but decreased in HUA mice with kidney injury (KI-HUA). In KI-HUA mice, treatment with anti-HUA agents, febuxostat, and benzbromarone decreased uric acid levels. However, febuxostat treatment also decreased URAT1 expression, whereas benzbromarone treatment increased its expression. Based on these in vivo findings, we propose that extracellular uric acid levels in the proximal tubule epithelial cells positively regulated URAT1 expression. In high uric acid cell models, URAT1 expression increased within 2 h of uric acid stimulation in a dose-dependent manner that supported our hypothesis. Therefore, our results suggest that URAT1 expression is positively regulated by the distinct extracellular uric acid levels in different HUA models.


Subject(s)
Hyperuricemia/metabolism , Organic Anion Transporters/metabolism , Animals , Benzbromarone/pharmacology , Disease Models, Animal , Febuxostat/pharmacology , HEK293 Cells , Humans , Hyperuricemia/drug therapy , Hyperuricemia/physiopathology , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Male , Mice, Inbred ICR , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Uric Acid/blood , Uric Acid/pharmacology
11.
Chemosphere ; 194: 43-48, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29197248

ABSTRACT

Microcystins are a family of cyclic heptapeptide toxins naturally produced by freshwater cyanobacteria. Microcystin-LR (MCLR) is believed to be the most toxic and common one with various pathological effects on human and mammals. However, the effects of MCLR on endothelial cells and vascular homeostasis have been largely unknown. We explored the mRNA and protein expression changes of several pro-inflammatory mediators in human umbilical vein endothelial cells (HUVECs) and C57BC/6 mice exposed to MCLR. Tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), especially interleukin-8 (IL-8) were remarkably upregulated both in endothelial cells and in serum. Increased endothelial permeability in vitro and chronic microvascular permeability in animals were also observed. Silencing the IL-8 gene with siRNA or blocking its cognate receptor, CXC-chemokine receptor type 2 (CXCR2), by a specific inhibitor efficiently prevented the MCLR induced leakage. These observations indicate a novel insight of inflammation triggered property of MCLR via IL-8/CXCR2 signaling, suggesting CXCR2 as a target molecule in protective strategy against the wide range pollution of microcystin.


Subject(s)
Capillary Permeability/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Interleukin-8/metabolism , Microcystins/pharmacology , Receptors, Interleukin-8B/metabolism , Animals , Cyanobacteria/metabolism , Cyanobacteria/pathogenicity , Humans , Inflammation/complications , Inflammation/etiology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/genetics , Marine Toxins , Mice , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
12.
J Cell Mol Med ; 20(6): 1139-49, 2016 06.
Article in English | MEDLINE | ID: mdl-26865248

ABSTRACT

Histone deacetylase (HDAC) 9, a member of class II HDACs, regulates a wide variety of normal and abnormal physiological functions, which is usually expressed at high levels in the brain and skeletal muscle. Although studies have highlighted the importance of HDAC-mediated epigenetic processes in the development of ischaemic stroke and very recent genome-wide association studies have identified a variant in HDAC9 associated with large-vessel ischemic stroke, the molecular events by which HDAC9 induces cerebral injury keep unclear. In this study, we found that HDAC9 was up-regulated in the ischaemic cerebral hemisphere after cerebral ischaemia/reperfusion (I/R) injury in rats and in vivo gene silencing of HDAC9 by recombinated lentivirus infection in the brain reduced cerebral injury in experimental stroke. We further demonstrated that HDAC9 contributed to oxygen-glucose deprivation-induced brain microvessel endothelial cell dysfunction as demonstrated by the increased inflammatory responses, cellular apoptosis and endothelial cell permeability dysfunction accompanied by reduced expression of tight-junction proteins. We further found that HDAC9 suppressed autophagy, which was associated with endothelial dysfunction. This study for the first time provides direct evidence that HDAC9 contributes to endothelial cell injury and demonstrates that HDAC9 is one of critical components of a signal transduction pathway that links cerebral injury to epigenetic modification in the brain.


Subject(s)
Brain Ischemia/complications , Brain Ischemia/enzymology , Brain/blood supply , Endothelial Cells/pathology , Histone Deacetylases/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Animals , Apoptosis , Autophagy , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Brain Ischemia/pathology , Cell Membrane Permeability , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gene Silencing , Gene Transfer Techniques , Glucose/deficiency , Histone Deacetylases/genetics , Inflammation/pathology , Lentivirus/metabolism , Male , Microvessels/pathology , Oxygen , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Up-Regulation
13.
Neurol Sci ; 36(11): 2027-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26169757

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive phospholipid that activates at least five known G-protein-coupled receptors (GPCRs): LPA1-LPA5. The nervous system is a major locus for LPA1 expression. LPA has been shown to regulate neuronal proliferation, migration, and differentiation during central nervous system development as well as neuronal survival. Furthermore, deficient LPA signaling has been implicated in several neurological disorders including neuropathic pain and schizophrenia. Parkinson's disease (PD) is a neurodegenerative movement disorder that results from the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). The specific molecular pathways that lead to DA neuron degeneration, however, are poorly understood. The influence of LPA in the differentiation of mesenchymal stem cells (MSCs) into DA neurons in vitro and LPA1 expression in a 6-hydroxydopamine (6-OHDA) lesion model of PD in vivo were examined in the present study. LPA induced neuronal differentiation in 80.2 % of the MSC population. These MSCs developed characteristic neuronal morphology and expressed the neuronal marker, neuron-specific enolase (NSE), while expression of the glial marker, glial fibrillary acidic protein (GFAP), was absent. Moreover, 27.6 % of differentiated MSCs were positive for tyrosine hydroxylase (TH), a marker for DA neurons. In the 6-OHDA PD rat model, LPA1 expression in the substantia nigra was significantly reduced compared to control. These results suggest LPA signaling via activation of LPA1 may be necessary for DA neuron development and survival. Furthermore, reduced LPA/LPA1 signaling may be involved in DA neuron degeneration thus contributing to the pathogenesis of PD.


Subject(s)
Dopaminergic Neurons/physiology , Lysophospholipids/metabolism , Neurogenesis/physiology , Parkinsonian Disorders/physiopathology , Receptors, Lysophosphatidic Acid/metabolism , Animals , Cell Survival/drug effects , Cell Survival/physiology , Central Nervous System Agents/administration & dosage , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Female , Glial Fibrillary Acidic Protein/metabolism , Lysophospholipids/administration & dosage , Male , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , Myenteric Plexus/metabolism , Neurogenesis/drug effects , Oxidopamine , Parkinsonian Disorders/pathology , Phosphopyruvate Hydratase/metabolism , Rats, Sprague-Dawley , Signal Transduction , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Tyrosine 3-Monooxygenase/metabolism
14.
Pharmazie ; 69(9): 688-93, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25272941

ABSTRACT

AIMS: To investigate the regulating effects of catalpol on the hypothalamic-pituitary- adrenocortical-axis (HPA) in an Alzheimer's disease (AD) rat model. METHODS: Healthy male Wistar Rats were selected. The AD model was generated by orthotopic injection of beta-amyloid 25-35 (Abeta25-35) into the right lateral ventricle. The animals were divided into five study groups: Catalpol at low dose (5 mg/kg), Catalpol at high dose (10 mg/kg), model control group and sham surgery control group, n = 9 respectively. The serum concentration of hydrocortisone (HYD), adrenocorticotropin (ACTH) and corticotropin releasing hormone (CRH) determined by Enzyme-Linked Immunosorbent Assay (ELISA). Structural alterations of the hypothalamus were examined by H&E stain and electron microscope. The CRH receptor 1 (CRHR1) positive neurons were detected with immunohistochemistry. RESULTS: Serum HYD level was significantly increased (p < 0.01), and both ACTH and CRH were dramatically decreased (p < 0.01) in the AD model group rats compared with normal control rats at day 7. Catalpol treatment was able to improve the hormone secretion disorder in AD model group rats compared with the model group (p < 0.01 or p < 0.05) in particular at 21 days. Structure damage of hypothalamus in the AD rat as evidenced less CRHR1 positive neurons, rough endoplasmic reticulum dilation and degranulation, and mitochondrial swelling under electron microscope. Catalpol treatment at both high and low doses was able to alleviate the structure damage of the hypothalamus in the AD rats. CONCLUSIONS: Catalpol could improve the endocrine function of the HPA and alleviate the structural damage of hypothalamus in AD rats.


Subject(s)
Alzheimer Disease/drug therapy , Hypothalamo-Hypophyseal System/drug effects , Iridoid Glucosides/pharmacology , Pituitary-Adrenal System/drug effects , Alzheimer Disease/physiopathology , Animals , Corticotropin-Releasing Hormone/blood , Enzyme-Linked Immunosorbent Assay , Hippocampus/pathology , Hypothalamo-Hypophyseal System/pathology , Hypothalamus/pathology , Immunohistochemistry , Microscopy, Electron, Transmission , Neurons/metabolism , Pituitary-Adrenal System/pathology , Rats , Receptors, Corticotropin-Releasing Hormone/biosynthesis
15.
Kidney Int ; 86(4): 712-25, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24717296

ABSTRACT

Studies have highlighted the importance of histone deacetylase (HDAC)-mediated epigenetic processes in the development of diabetic complications. Inhibitors of HDAC are a novel class of therapeutic agents in diabetic nephropathy, but currently available inhibitors are mostly nonselective inhibit multiple HDACs, and different HDACs serve very distinct functions. Therefore, it is essential to determine the role of individual HDACs in diabetic nephropathy and develop HDAC inhibitors with improved specificity. First, we identified the expression patterns of HDACs and found that, among zinc-dependent HDACs, HDAC2/4/5 were upregulated in the kidney from streptozotocin-induced diabetic rats, diabetic db/db mice, and in kidney biopsies from diabetic patients. Podocytes treated with high glucose, advanced glycation end products, or transforming growth factor-ß (common detrimental factors in diabetic nephropathy) selectively increased HDAC4 expression. The role of HDAC4 was evaluated by in vivo gene silencing by intrarenal lentiviral gene delivery and found to reduce renal injury in diabetic rats. Podocyte injury was associated with suppressing autophagy and exacerbating inflammation by HDAC4-STAT1 signaling in vitro. Thus, HDAC4 contributes to podocyte injury and is one of critical components of a signal transduction pathway that links renal injury to autophagy in diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Histone Deacetylases/metabolism , Podocytes/metabolism , Podocytes/pathology , Repressor Proteins/metabolism , Animals , Autophagy , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Enzyme Inhibitors/pharmacology , Gene Silencing , Glucose/pharmacology , Glycation End Products, Advanced/pharmacology , Histone Deacetylase 2/metabolism , Histone Deacetylases/analysis , Histone Deacetylases/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Podocytes/chemistry , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Repressor Proteins/analysis , Repressor Proteins/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , Signal Transduction , Transforming Growth Factor beta/pharmacology , Up-Regulation
16.
J Cereb Blood Flow Metab ; 34(4): 660-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424382

ABSTRACT

Although the innate immune response to induce postischemic inflammation is considered as an essential step in the progression of cerebral ischemia injury, the role of innate immunity mediator NLRP3 in the pathogenesis of ischemic stroke is unknown. In this study, focal ischemia was induced by middle cerebral artery occlusion in NLRP3(-/-), NOX2(-/-), or wild-type (WT) mice. By magnetic resonance imaging (MRI), Evans blue permeability, and electron microscopic analyses, we found that NLRP3 deficiency ameliorated cerebral injury in mice after ischemic stroke by reducing infarcts and blood-brain barrier (BBB) damage. We further showed that the contribution of NLRP3 to neurovascular damage was associated with an autocrine/paracrine pattern of NLRP3-mediated interleukin-1ß (IL-1ß) release as evidenced by increased brain microvessel endothelial cell permeability and microglia-mediated neurotoxicity. Finally, we found that NOX2 deficiency improved outcomes after ischemic stroke by mediating NLRP3 signaling. This study for the first time shows the contribution of NLRP3 to neurovascular damage and provides direct evidence that NLRP3 as an important target molecule links NOX2-mediated oxidative stress to neurovascular damage in ischemic stroke. Pharmacological targeting of NLRP3-mediated inflammatory response at multiple levels may help design a new approach to develop therapeutic strategies for prevention of deterioration of cerebral function and for the treatment of stroke.


Subject(s)
Blood-Brain Barrier/pathology , Carrier Proteins/metabolism , Ischemic Attack, Transient/pathology , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Stroke/pathology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/metabolism , Carrier Proteins/genetics , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glucose/metabolism , Ischemic Attack, Transient/metabolism , Magnetic Resonance Imaging , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Neurons/metabolism , Neurons/pathology , Oxidative Stress/physiology , Oxygen/metabolism , Signal Transduction , Stroke/metabolism
17.
Channels (Austin) ; 8(1): 13-9, 2014.
Article in English | MEDLINE | ID: mdl-24299922

ABSTRACT

An increasing number of studies have implicated that the activation of innate immune system and inflammatory mechanisms are of importance in the pathogenesis of numerous diseases. The innate immune system is present in almost all multicellular organisms in response to pathogens or tissue injury, which is performed via germ-line encoded pattern-recognition receptors (PRRs) to recognize pathogen-associated molecular patterns (PAMPs) or dangers-associated molecular patterns (DAMPs). Intracellular pathways linking immune and inflammatory response to ion channel expression and function have been recently identified. Among ion channels, transient receptor potential (TRP) channels are a major family of non-selective cation-permeable channels that function as polymodal cellular sensors involved in many physiological and pathological processes. In this review, we summarize current knowledge about classifications, functions, and interactions of TRP channels and PRRs, which may provide new insights into their roles in the pathogenesis of inflammatory diseases.


Subject(s)
Inflammation/metabolism , Receptors, Pattern Recognition/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Calcium Signaling , Humans , Immunity, Innate , Inflammation/immunology , Receptors, Pattern Recognition/immunology , Transient Receptor Potential Channels/immunology
18.
Hypertension ; 62(3): 506-11, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23856489

ABSTRACT

Although hyperhomocysteinemia (hHcys) has been recognized as an important independent risk factor in the progression of end-stage renal disease and in the development of cardiovascular complications related to end-stage renal disease, the mechanisms triggering the pathogenic actions of hHcys are not yet fully understood. The present study was designed to investigate the contribution of nucleotide-binding oligomerization domain containing 2 (NOD2), an intracellular innate immunity mediator, to the development of glomerulosclerosis in hHcys. Our results showed that NOD2 deficiency ameliorated renal injury in mice with hHcys. We further discovered the novel role of NOD2 in mediating Ca(2+) signaling and found that homocysteine-induced NOD2 expression enhanced transient receptor potential cation channel 6 (TRPC6) expression and TRPC6-mediated calcium influx and currents, leading to intracellular Ca(2+) release, ultimately resulting in podocyte cytoskeleton rearrangement and apoptosis. Moreover, we found that nephrin expression was downregulated dependently by NOD2, and overexpression of nephrin attenuated homocysteine-induced TRPC6 expression in podocytes. The results add evidence to support the essential role of nephrin in mediating NOD2-induced TRPC6 expression in hHcys. In conclusion, our results for the first time establish a previously unknown function of NOD2 for the regulation of TRPC6 channels, suggesting that TRPC6-dependent Ca(2+) signaling is one of the critical signal transduction pathways that links innate immunity mediator NOD2 to podocyte injury. Pharmacological targeting of NOD2 signaling pathways at multiple levels may help design a new approach to develop therapeutic strategies for treatment of hHcys-associated end-stage renal disease.


Subject(s)
Calcium Signaling/physiology , Hyperhomocysteinemia/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Podocytes/metabolism , TRPC Cation Channels/metabolism , Animals , Cytoskeleton , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/pathology , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Podocytes/pathology , TRPC Cation Channels/genetics , TRPC6 Cation Channel
19.
Kidney Int ; 84(2): 265-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23594678

ABSTRACT

An increasing number of clinical and animal model studies indicate that activation of the innate immune system and inflammatory mechanisms are important in the pathogenesis of diabetic nephropathy. Nucleotide-binding oligomerization domain containing 2 (NOD2), a member of the NOD-like receptor family, plays an important role in innate immune response. Here we explore the contribution of NOD2 to the pathogenesis of diabetic nephropathy and found that it was upregulated in kidney biopsies from diabetic patients and high-fat diet/streptozotocin-induced diabetic mice. Further, NOD2 deficiency ameliorated renal injury in diabetic mice. In vitro, NOD2 induced proinflammatory response and impaired insulin signaling and insulin-induced glucose uptake in podocytes. Moreover, podocytes treated with high glucose, advanced glycation end-products, tumor necrosis factor-α, or transforming growth factor-ß (common detrimental factors in diabetic nephropathy) significantly increased NOD2 expression. NOD2 knockout diabetic mice were protected from the hyperglycemia-induced reduction in nephrin expression. Further, knockdown of NOD2 expression attenuated high glucose-induced nephrin downregulation in vitro, supporting an essential role of NOD2 in mediating hyperglycemia-induced podocyte dysfunction. Thus, NOD2 is one of the critical components of a signal transduction pathway that links renal injury to inflammation and podocyte insulin resistance in diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Inflammation Mediators/metabolism , Insulin Resistance , Nephritis/etiology , Nod2 Signaling Adaptor Protein/metabolism , Podocytes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/blood , Biomarkers/urine , Blood Glucose/metabolism , Cells, Cultured , Creatinine/blood , Creatinine/urine , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/immunology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/prevention & control , Diet, High-Fat , Female , Glucose Transporter Type 4/metabolism , Humans , Insulin/blood , Lipids/blood , MAP Kinase Signaling System , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Middle Aged , Nephritis/genetics , Nephritis/immunology , Nephritis/metabolism , Nephritis/pathology , Nephritis/prevention & control , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Podocytes/pathology , Time Factors , Up-Regulation
20.
Exp Ther Med ; 4(4): 728-732, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23170134

ABSTRACT

The aim of this study was to examine the association between the -149C>T polymorphism of DNA methyltransferase 3B (DNMT3B) and colorectal cancer (CRC) susceptibility. A comprehensive search was conducted to identify all case-control studies of the -149C>T polymorphism of DNMT3B and CRC risk. Statistical analysis was performed with the software program Stata (version 12.0) and Review Manager (version 5.0). A total of seven eligible studies, including 2,666 cases and 4,022 controls, associating the DNMT3B polymorphism of -149C>T with the risk of CRC were identified. These studies suggested no significant associations between the -149C>T polymorphism of the DNMT3B gene and the risk of developing CRC in the recessive, dominant and co-dominant models [for CC vs. TT: odds ratio (OR), 0.90; 95% confidence interval (CI), 0.90-1.25; P=0.37; for the recessive model: OR, 0.54, 95% CI, 0.28-1.04; P<0.00001; for the dominant model: OR, 1.07; 95% CI, 0.93-1.23; P=0.83 and C allele vs. T allele: OR, 0.70; 95% CI, 0.43-1.13; P<0.00001]. In the subgroup analysis, no significant associations were found in the European populations (for CC vs. TT: OR, 1.09; 95% CI, 0.92-1.30; P=0.88; for the recessive model: OR, 1.00; 95% CI, 0.88-1.13; P=0.14; for the dominant model: OR, 1.50; 95% CI, 0.89-2.54; P<0.00001 and C allele vs. T allele: OR, 0.70; 95% CI, 0.38-1.28; P<0.00001). No significant association was found between the -149C>T polymorphism in DNMT3B and CRC susceptibility.

SELECTION OF CITATIONS
SEARCH DETAIL
...