Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Int Immunopharmacol ; 99: 107901, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34273637

ABSTRACT

Periodontitis is initiated by serious and sustained bacterial infection and ultimately results in chronic immune-mediated inflammation, tissue destruction, and bone loss. The pathogenesis of periodontitis remains unclear. Host immunological responses to periodontal bacteria ultimately determine the severity and mechanisms governing periodontitis progression. This study aimed to clarify the effect of the hypoxia-inducible factor-1α (HIF-1α) activator dimethyloxalylglycine (DMOG) on a mouse periodontitis model and its underlying role in macrophage polarization. qRT-PCR analysis showed that DMOG inhibited the M1-like polarization of both RAW264.7 macrophages and murine bone marrow macrophages (BMMs) and downregulated TNF-α, IL-6, CD86, and MCP-1 expression in vitro. Immunofluorescence staining and flow cytometry also confirmed the less percentage of F4/80 + CD86 + cells after DMOG treatment. The phosphorylation of NF-κB pathway was also inhibited by DMOG with higher level of HIF-1α expression. Furthermore, mice treated with DMOG showed decreased alveolar bone resorption in the experimental periodontitis model, with significant increases in alveolar bone volume/tissue volume (BV/TV) and bone mineral density (BMD). DMOG treatment of mice decreased the ratio of M1/M2 (CD86+/CD206+) macrophages in periodontal tissues, resulting in the downregulation of proinflammatory cytokines such as TNF-α and IL-6 and increased levels of anti-inflammatory factors such as IL-4 and IL-10. DMOG treatment promoted the number of HIF-1α-positive cells in periodontal tissues. This study demonstrated the cell-specific roles of DMOG in macrophage polarization in vitro and provided insight into the mechanism underlying the protective effect of DMOG in a model of periodontitis.


Subject(s)
Alveolar Bone Loss/drug therapy , Amino Acids, Dicarboxylic/therapeutic use , Macrophages/drug effects , Periodontitis/drug therapy , Alveolar Bone Loss/diagnostic imaging , Alveolar Bone Loss/immunology , Alveolar Bone Loss/pathology , Amino Acids, Dicarboxylic/pharmacology , Animals , Cytokines/genetics , Hypoxia-Inducible Factor 1, alpha Subunit , Macrophages/immunology , Male , Maxilla/diagnostic imaging , Maxilla/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , Periodontitis/diagnostic imaging , Periodontitis/immunology , Periodontitis/pathology , RAW 264.7 Cells , Signal Transduction/drug effects , X-Ray Microtomography
2.
Exp Neurol ; 334: 113436, 2020 12.
Article in English | MEDLINE | ID: mdl-32814068

ABSTRACT

Despite the ability of peripheral nerves to regenerate after injury, failure occurs due to an inability of supporting cells to maintain growth, resulting in long-term consequences such as sensorimotor dysfunction and neuropathic pain. Here, we investigate the potential of engaging the cellular adaptive response to hypoxia, via inhibiting its negative regulators, to enhance the regenerative process. Under normoxic conditions, prolyl hydroxylase domain (PHD) proteins 1, 2, and 3 hydroxylate the key metabolic regulator hypoxia inducible factor 1α (HIF1α), marking it for subsequent proteasomal degradation. We inhibited PHD protein function systemically via either individual genetic deletion or pharmacological pan-PHD inhibition using dimethyloxalylglycine (DMOG). We show enhanced axonal regeneration after sciatic nerve crush injury in PHD1-/- mice, PHD3-/- mice, and in DMOG-treated mice, and in PHD1-/- and DMOG-treated mice a reduction in hypersensitivity to cooling after permanent sciatic ligation. Electromyographically, PHD1-/- and PHD3-/- mice showed an increased CMAP amplitude one-month post-injury, probably due to protection against denervation induced muscle atrophy, while DMOG-treated and PHD2+/- mice showed reduced latencies, indicating improved motor axon function. DMOG treatment did not affect the growth of dorsal root ganglion neurites in vitro, suggesting a lack of direct effects of DMOG on axonal regrowth. Enhanced regeneration in vivo was concurrent with an increase in macrophage density, and a shift in macrophage polarization state ratios (from M1-like toward M2-like) in DMOG-treated animals. These results indicate PHD proteins as a novel therapeutic target to improve regenerative and functional outcomes after peripheral nerve injury without manipulating molecular O2.


Subject(s)
Axons/physiology , Hypoxia/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Recovery of Function/physiology , Amino Acids, Dicarboxylic/pharmacology , Amino Acids, Dicarboxylic/therapeutic use , Animals , Axons/drug effects , Cells, Cultured , Hypoxia/drug therapy , Hypoxia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/pathology , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Prolyl-Hydroxylase Inhibitors/therapeutic use , Recovery of Function/drug effects
3.
J Neurotrauma ; 36(24): 3394-3409, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31232175

ABSTRACT

Spinal cord injury (SCI) is a devastating neurological disorder that usually leads to a loss of motor and sensory function in patients. The expression of hypoxia inducible factor-1α (HIF-1α) is increased, and exerts a protective role after traumatic SCI. However, the endogenous activity of HIF-1α is insufficient for promoting functional recovery. The present study tested the potential effect of the sustained activation of HIF-1α by the prolylhydroxylase (PHD) inhibitor dimethyloxalylglycine (DMOG) on anti-apoptotic process and the regulation of axonal regeneration after SCI. Here, we found that treatment with DMOG significantly increased the expression of HIF-1α and that the stabilization of HIF-1α induced by DMOG not only decreased the expression of apoptotic proteins to promote neural survival, but also enhanced axonal regeneration by regulating microtubule stabilization in vivo and in vitro. In addition, we found that DMOG promoted neural survival and axonal regeneration by activating autophagy, which is induced by the HIF-1α/BNIP3 signaling pathway, and that the inhibition of HIF-1α or autophagy abrogated the protective effect of DMOG, as expected. Taken together, our results demonstrate that treatment with DMOG improves functional recovery after SCI and that DMOG may serve as a potential candidate for treating SCI.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Axons/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nerve Regeneration/physiology , Recovery of Function/physiology , Spinal Cord Injuries/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Axons/drug effects , Female , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Nerve Regeneration/drug effects , PC12 Cells , Protein Stability/drug effects , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/physiopathology
4.
Physiol Behav ; 199: 265-272, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30500334

ABSTRACT

Mesenchymal stem cell (MSC) transplantation therapy has been proposed as a promising approach for the treatment of neurodegenerative disease. Chemical and pharmacological preconditioning before transplantation could optimize the therapeutic properties of transplanted MSCs. In this study, we hypothesized that preconditioning treatment with a prolyl hydroxylase inhibitor, dimethyloxalylglycine (DMOG), will increase MSC efficacy and paracrine effects in an amyloid-ß (Aß)-injected Alzheimer rat model. MSCs were incubated in different concentrations of DMOG for 24 h. Cell viability, migration, and antioxidant capacity was assessed in DMOG-treated and non-treated MSCs before transplantation into Aß-injected rats. In vitro analysis revealed that DMOG treatment increased cell viability, migration, and expression of CXCR4, CCR2, Nrf2, and HIF-1α in the MSCs. Our in vivo results show that DMOG preconditioning enhances a MSC-mediated rescue of learning and memory function in Aß-injected rats. Furthermore, we found an increased level of BDNF and total antioxidant capacity in the hippocampus of Aß-injected rats following transplantation of preconditioned relative to untreated MSCs. Our results suggest that preconditioning MSCs with DMOG before transplantation may enhance the efficacy of stem cell based therapy in neurodegenerative disease.


Subject(s)
Alzheimer Disease/therapy , Amino Acids, Dicarboxylic/therapeutic use , Cell Survival/drug effects , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Alzheimer Disease/chemically induced , Amino Acids, Dicarboxylic/pharmacology , Amyloid beta-Peptides , Animals , Disease Models, Animal
5.
Allergy ; 74(4): 753-766, 2019 04.
Article in English | MEDLINE | ID: mdl-30394557

ABSTRACT

BACKGROUND: When an immune cell migrates from the bloodstream to a site of chronic inflammation, it experiences a profound decrease in microenvironmental oxygen levels leading to a state of cellular hypoxia. The hypoxia-inducible factor-1α (HIF-1α) promotes an adaptive transcriptional response to hypoxia and as such is a major regulator of immune cell survival and function. HIF hydroxylases are the family of oxygen-sensing enzymes primarily responsible for conferring oxygen dependence upon the HIF pathway. METHODS: Using a mouse model of allergic contact dermatitis (ACD), we tested the effects of treatment with the pharmacologic hydroxylase inhibitor DMOG, which mimics hypoxia, on disease development. RESULTS: Re-exposure of sensitized mice to 2,4-dinitrofluorobenzene (DNFB) elicited inflammation, edema, chemokine synthesis (including CXCL1 and CCL5) and the recruitment of neutrophils and eosinophils. Intraperitoneal or topical application of the pharmacologic hydroxylase inhibitors dymethyloxalylglycine (DMOG) or JNJ1935 attenuated this inflammatory response. Reduced inflammation was associated with diminished recruitment of neutrophils and eosinophils but not lymphocytes. Finally, hydroxylase inhibition reduced cytokine-induced chemokine production in cultured primary keratinocytes through attenuation of the JNK pathway. CONCLUSION: These data demonstrate that hydroxylase inhibition attenuates the recruitment of neutrophils to inflamed skin through reduction of chemokine production and increased neutrophilic apoptosis. Thus, pharmacologic inhibition of HIF hydroxylases may be an effective new therapeutic approach in allergic skin inflammation.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Dermatitis, Allergic Contact/prevention & control , Mixed Function Oxygenases/antagonists & inhibitors , Amino Acids, Dicarboxylic/pharmacology , Animals , Cell Movement/drug effects , Cytokines/metabolism , Eosinophils/cytology , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation/drug therapy , Mice , Neutrophils/cytology
6.
Int J Cardiol ; 272: 281-287, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30177233

ABSTRACT

BACKGROUND: Diastolic dysfunction is emerging as a leading cause of heart failure in aging population. Induction of hypoxia tolerance and reprogrammed cell metabolism have emerged as novel therapeutic strategies for the treatment of cardiovascular diseases. METHODS AND RESULTS: In the present study, we showed that deletion of sirtuin 3 (SIRT3) resulted in a diastolic dysfunction together with a significant increase in the expression of prolyl hydroxylases (PHD) 1 and 2. We further investigated the involvement of PHD in the development of diastolic dysfunction by treating the 12-14 months old mice with a PHD inhibitor, dimethyloxalylglycine (DMOG) for 2 weeks. DMOG treatment increased the expression of hypoxia-inducible factor (HIF)-1α in the endothelium of coronary arteries. This was accompanied by a significant improvement of coronary flow reserve and diastolic function. Inhibition of PHD altered endothelial metabolism by increasing glycolysis and reducing oxygen consumption. Most importantly, treatment with DMOG completely reversed the pre-existing diastolic dysfunction in the endothelial-specific SIRT3 deficient mice. CONCLUSIONS: Our findings demonstrate that inhibition of PHD and reprogrammed cell metabolism can reverse the pre-existed diastolic dysfunction in SIRT3 deficient mice. Our study provides a potential therapeutic strategy of induction of hypoxia tolerance for patients with diastolic dysfunction associated with coronary microvascular dysfunction, especially in the aging population with reduced SIRT3.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Endothelium, Vascular/metabolism , Heart Failure, Diastolic/drug therapy , Heart Failure, Diastolic/metabolism , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/therapeutic use , Amino Acids, Dicarboxylic/pharmacology , Animals , Diastole/drug effects , Diastole/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Male , Mice , Mice, Knockout , Mice, Transgenic , Prolyl-Hydroxylase Inhibitors/pharmacology , Sirtuin 3/deficiency
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 152-164, 2017 01.
Article in English | MEDLINE | ID: mdl-27664837

ABSTRACT

Cerebral ischemic stroke is one of the leading causes of death and disability worldwide. Therapeutic interventions to minimize ischemia-induced neural damage are limited due to poor understanding of molecular mechanisms mediating complex pathophysiology in stroke. Recently, epigenetic mechanisms mostly histone lysine (K) acetylation and deacetylation have been implicated in ischemic brain damage and have expanded the dimensions of potential therapeutic intervention to the systemic/local administration of histone deacetylase inhibitors. However, the role of other epigenetic mechanisms such as histone lysine methylation and demethylation in stroke-induced damage and subsequent recovery process is elusive. Here, we established an Internal Carotid Artery Occlusion (ICAO) model in CD1 mouse that resulted in mild to moderate level of ischemic damage to the striatum, as suggested by magnetic resonance imaging (MRI), TUNEL and histopathological staining along with an evaluation of neurological deficit score (NDS), grip strength and rotarod performance. The molecular investigations show dysregulation of a number of histone lysine methylases (KMTs) and few of histone lysine demethylases (KDMs) post-ICAO with significant global attenuation in the transcriptionally repressive epigenetic mark H3K9me2 in the striatum. Administration of Dimethyloxalylglycine (DMOG), an inhibitor of KDM4 or JMJD2 class of histone lysine demethylases, significantly ameliorated stroke-induced NDS by restoring perturbed H3K9me2 levels in the ischemia-affected striatum. Overall, these results highlight the novel role of epigenetic regulatory mechanisms controlling the epigenetic mark H3K9me2 in mediating the stroke-induced striatal damage and subsequent repair following mild to moderate cerebral ischemia.


Subject(s)
Brain Ischemia/genetics , Epigenesis, Genetic , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Lysine/genetics , Amino Acids, Dicarboxylic/pharmacology , Amino Acids, Dicarboxylic/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Demethylation/drug effects , Epigenesis, Genetic/drug effects , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Male , Methylation/drug effects , Mice
8.
J Cardiovasc Pharmacol ; 67(1): 68-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26485211

ABSTRACT

BACKGROUND: One important determinant of longevity in congenital heart disease is right ventricular (RV) function, and this is especially true in cyanotic congenital heart disease. However, there is a paucity of data concerning right ventricular remodeling (RVR) in the setting of chronic hypoxia. Dimethyloxalylglycine (DMOG) is a competitive inhibitor of hypoxia-inducible factor (HIF)-hydroxylated prolyl hydroxylase and has been shown to play an important role against ischemia-reperfusion myocardial injury. METHODS: We tested the hypothesis that DMOG prevents the development RVR after chronic hypoxia exposure. Rats were injected with saline or DMOG and exposed to room air or continued hypoxia for 4 weeks. In addition, we explored the response of myocardial erythropoietin and its receptor to hypoxic exposure. RESULTS: Treatment with DMOG attenuated myocardial fibrosis, apoptosis, and oxidative stress, which lead to enhanced RV contractile function. As an endpoint of HIF-dependent cardioprotection, a novel pathway in which nuclear factor kappa B links HIF-1 transcription was defined. CONCLUSIONS: This study supports a role for HIF-1 stabilizers in the treatment of RVR and brings into question the commonly held concept that RVR follows a linear relationship with increased RV afterload.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/drug therapy , Hypoxia/metabolism , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects , Amino Acids, Dicarboxylic/pharmacology , Animals , Chronic Disease , Male , Rats , Rats, Sprague-Dawley , Ventricular Function, Right/physiology , Ventricular Remodeling/physiology
9.
J Heart Lung Transplant ; 35(1): 99-107, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26255815

ABSTRACT

OBJECTIVE: Hypoxia inducible factor (HIF)-1 pathway signalling has a protective effect against ischemia/reperfusion injury. The prolyl-hydroxylase inhibitor dimethyloxalylglycine (DMOG) activates the HIF-1 pathway by stabilizing HIF-1α. In a rat model of brain death (BD)-associated donor heart dysfunction we tested the hypothesis that pre-treatment of brain-dead donors with DMOG would result in a better graft heart condition. METHODS: BD was induced in anesthetized Lewis rats by inflating a subdurally placed balloon catheter. Controls underwent sham operations. Then, rats were injected with an intravenous dose of DMOG (30 mg/kg) or an equal volume of physiologic saline. After 5 hours of BD or sham operation, hearts were perfused with a cold (4°C) preservation solution (Custodiol; Dr. Franz Köhler Chemie GmbH; Germany), explanted, stored at 4°C in Custodiol, and heterotopically transplanted. Graft function was evaluated 1.5 hours after transplantation. RESULTS: Compared with control, BD was associated with decreased left ventricular systolic and diastolic function. DMOG treatment after BD improved contractility (end-systolic pressure volume relationship E'max: 3.7 ± 0.6 vs 3.1 ± 0.5 mm Hg/µ1; p < 0.05) and left ventricular stiffness (end-diastolic pressure volume relationship: 0.13 ± 0.03 vs 0.31 ± 0.06 mm Hg/µ1; p < 0.05) 5 hours later compared with the brain-dead group. After heart transplantation, DMOG treatment of brain-dead donors significantly improved the altered systolic function and decreased inflammatory infiltration, cardiomyocyte necrosis, and DNA strand breakage. In addition, compared with the brain-dead group, DMOG treatment moderated the pro-apoptotic changes in the gene and protein expression. CONCLUSIONS: In a rat model of potential brain-dead heart donors, pre-treatment with DMOG resulted in improved early recovery of graft function after transplantation. These results support the hypothesis that activation of the HIF-1 pathway has a protective role against BD-associated cardiac dysfunction.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Heart Transplantation , Reperfusion Injury/drug therapy , Tissue Donors , Ventricular Function, Left/drug effects , Animals , Brain/blood supply , Disease Models, Animal , Male , Rats , Rats, Inbred Lew , Reperfusion Injury/physiopathology , Ventricular Function, Left/physiology
10.
J Control Release ; 217: 221-7, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26385165

ABSTRACT

Targeting hypoxia-sensitive pathways has recently been proposed as a new therapeutic approach to the treatment of intestinal inflammation. HIF-hydroxylases are enzymes which confer hypoxic-sensitivity upon the hypoxia-inducible factor (HIF), a major regulator of the adaptive response to hypoxia. Previous studies have shown that systemic (intraperitoneal) administration of hydroxylase inhibitors such as dimethyloxalylglycine (DMOG) is profoundly protective in multiple models of colitis, however the therapeutic potential of this approach is limited due to potential side-effects associated with systemic drug exposure and the fact that orally delivered DMOG is ineffective (likely due to drug inactivation by gastric acid). In order to overcome these issues, we formulated DMOG in a liquid emulsion drug delivery system which, when coated with specific polymer coatings, permits oral delivery of a reduced dose which is released locally throughout the colon. This colon-targeted DMOG formulation demonstrated increased relative colonic bioactivity with reduced systemic exposure and provided a similar degree of protection to systemic (intraperitoneal) administration at a 40-fold lower dose in DSS-induced colitis. In summary, targeted delivery of DMOG to the colon provides local protection resulting in enhanced efficacy with reduced systemic exposure in the treatment of colitis. This novel approach to targeting hydroxylase inhibitors to specific diseased regions of the GI tract may improve it's potential as a new therapeutic in inflammatory bowel diseases such as ulcerative colitis.


Subject(s)
Amino Acids, Dicarboxylic/administration & dosage , Colitis/drug therapy , Mixed Function Oxygenases/antagonists & inhibitors , Administration, Oral , Amino Acids, Dicarboxylic/therapeutic use , Animals , Colitis/chemically induced , Colon/metabolism , Dextran Sulfate , Disease Models, Animal , Drug Delivery Systems , Female , HeLa Cells , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Mice , Mice, Transgenic , NF-kappa B/metabolism , Treatment Outcome
11.
BMC Oral Health ; 15: 60, 2015 May 16.
Article in English | MEDLINE | ID: mdl-25981588

ABSTRACT

BACKGROUND: Rapid wound healing of oral soft tissue may reduce the opportunity of infection and discomfort of patients. Previous studies have demonstrated that enhancement of angiogenesis is an effective way to accelerate wound repair. In this study, to enhance angiogenesis and healing of palatal wounds, dimethyloxalylglycine (DMOG) was applied to a rat palatal wound model. DMOG is known to inhibit oxygen-dependent degradation of hypoxia inducible factor-1 alpha (HIF-1α), which can lead to up-regulation of angiogenesis markers, favoring wound repair. We also evaluated the effects of DMOG on cell migration and HIF-1α expression of rat palatal (RP) cells. Furthermore, mRNA and protein expression of vascular endothelial growth factor (VEGF) were analyzed in DMOG-treated RP cells. METHODS: Primary cultures of rat palatal (RP) cells were obtained from Sprague-Dawley (SD) rats. Effects of DMOG on cell viability and migration of RP cells were evaluated by using a formazan and culture insert, respectively. VEGF mRNA was observed by real-time PCR, and VEGF and HIF-1α proteins were detected by Western blotting. For the animal study, excisional wounds, 3 mm in diameter, were made at the central part of the palate of SD rats. DMOG with hyaluronic acid ointment was topically applied three times during 1 week, and then wound closures were quantitated photographically and histologically. RESULTS: DMOG was cytotoxic to RP cells at concentrations higher than 2 mM and did not affect cell migration at non-cytotoxic concentrations. mRNA and protein expression of VEGF were significantly stimulated by DMOG treatment. The protein level of HIF-1α was also stabilized in RP cells by DMOG. In the animal study, groups treated with 1 mg/ml DMOG showed an increase of rat palatal wound contractures. CONCLUSIONS: DMOG enhanced wound healing of rat palatal mucosa, which was likely due to the angiogenic effect of the agent.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Mouth Mucosa/injuries , Palate/injuries , Angiogenic Proteins/pharmacology , Animals , Cell Culture Techniques , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Hyaluronic Acid/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Male , Models, Animal , Mouth Mucosa/drug effects , Palate/drug effects , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/drug effects , Wound Healing/drug effects
12.
Acta Biochim Biophys Sin (Shanghai) ; 46(2): 112-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24389644

ABSTRACT

Hyperlipidemia is an independent risk factor in the development of ischemic heart disease, which can increase myocardial susceptibility to ischemia/reperfusion (I/R) injury. Ischemic postconditioning (PostC) has now been demonstrated as a novel strategy to harness nature's protection against myocardial I/R injury in normal conditions. However, the effect of PostC on hyperlipidemic animals remains elusive. It has been shown in our previous study that PostC reduces the myocardial I/R injury, and hypoxia-inducible factor-1α (HIF-1α) may play an important role in the cardioprotective mechanisms of PostC on normal rats. Here, we tested the hypothesis that the cardioprotection of PostC on hyperlipidemic rats is associated with the up-regulated HIF-1α expression. Male Wistar rats were fed with a high-fat diet for 8 weeks, and then randomly divided into five groups: sham, I/R, dimethyloxalylglycine (DMOG) + I/R, PostC, and DMOG + PostC group. The detrimental indices induced by I/R injury included infarct size, plasma creatine kinase (CK) activity and caspase-3 activity. The results showed that PostC could reduce the infarct size, when compared with the I/R group, which was consistent with the significant lower levels of plasma CK activity and caspase-3 activity, and that it increased the expression of HIF-1α in hyperlipidemic rats. When DMOG was given before PostC to up-regulate HIF-1α protein level, the degree of I/R injury was attenuated. In conclusion, these data suggested that the up-regulation of HIF-1α may be one of the cardioprotective mechanisms of PostC against I/R injury in hyperlipidemic rats.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Ischemic Postconditioning , Myocardial Reperfusion Injury/therapy , Amino Acids, Dicarboxylic/therapeutic use , Animals , Diet, High-Fat , Hyperlipidemias/complications , Male , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Rats , Rats, Wistar , Up-Regulation
13.
Magn Reson Med ; 70(5): 1481-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23280475

ABSTRACT

MRI is a method of choice for assessing anatomical structures or angiogenesis-related parameters noninvasively during tumor progression. Typically, tumor tissue displays a high degree of heterogeneity that can be evaluated using pattern analysis (PA), which comprises shape and texture analysis. This work aims at implementing PA methods to study angiogenesis in a murine tumor model and testing its sensitivity with regard to detecting changes elicited by administration of a drug. Twelve balb/c-nude mice were injected subcutaneously with 10(6) C51 cells (colon carcinoma). A first group (N = 6) of animals was treated with dimethyloxalylglycine, a drug known to stabilize hypoxia-inducible-factor-α, which among other functions, is involved in angiogenesis. The second group (N = 6) was treated with saline. MRI experiments assessing tumor blood volume and permeability-maps (K(trans) ) were performed immediately before and 6 days after drug treatment. Data have been analyzed using standard histogram analysis and PA. Standard histogram analysis did not reveal any difference between the two groups, neither before nor after the treatment. In contrast, PA revealed significant differences between drug and placebo treated mice in the texture of the TBV and K(trans) maps after drug treatment, but not with regard to tumors shapes. The results indicated that in view of the heterogeneity of tumor tissue, standard histogram analysis appears insensitive in picking-up differences in response to treatment, while PA appears to be particularly sensitive to changes in texture.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Magnetic Resonance Imaging/methods , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Pattern Recognition, Automated/methods , Algorithms , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Colonic Neoplasms/complications , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/complications , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome , Tumor Burden
14.
Inflamm Bowel Dis ; 17(10): 2058-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21910167

ABSTRACT

BACKGROUND: This study investigates whether deoxy-2-[18F]fluoro-d-glucose (FDG) micro-positron emission tomography (µPET)/computed tomography (CT) can serve as a tool for monitoring of the commonly used dextran sodium sulfate (DSS)-induced murine model of inflammatory bowel disease (IBD). METHODS: DSS-colitis was induced in Sv129 mice. In a first experiment, four animals were serially scanned with CT and FDG-µPET on days 0, 3, 7, 11, and 14. The ratio of the mean voxel count of the PET images in the colon and the brain was compared with the histological inflammation score and the colonic myeloperoxidase levels. A second experiment was performed to investigate whether FDG-µPET was able to detect differences in inflammation between two DSS-treated groups, one receiving placebo (n = 4) and one receiving dimethyloxalylglycine (DMOG) (n = 4), a compound that protects against DSS-induced colitis. RESULTS: The progression of the colonic/brain FDG-signal ratio (over days 0-14) agreed with the predicted histological inflammation score, obtained from a parallel DSS-experiment. Moreover, the quantification of normalized colonic FDG-activity at the final timepoint (day 14) showed an excellent correlation with both the MPO levels (Spearman's rho = 1) and the histological inflammation score (Spearman's rho = 0.949) of the scanned mice. The protective action of DMOG in DSS colitis was clearly demonstrated with FDG-µPET/CT (normalized colonic FDG-activity DMOG versus placebo: P < 0.05). CONCLUSIONS: FDG-µPET-CT is a feasible and reliable noninvasive method to monitor murine DSS-induced colitis. The implementation of this technique in this widely used IBD model opens a new window for pathophysiological research and high-throughput screening of potential therapeutic compounds in preclinical IBD research.


Subject(s)
Colitis/immunology , Colitis/pathology , Dextran Sulfate/toxicity , Inflammation/immunology , Inflammation/pathology , Positron-Emission Tomography , Tomography, X-Ray Computed , Amino Acids, Dicarboxylic/therapeutic use , Animals , Colitis/chemically induced , Colitis/drug therapy , Disease Models, Animal , Female , Image Processing, Computer-Assisted , Inflammation/chemically induced , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(3): 322-5, 2011 May.
Article in Chinese | MEDLINE | ID: mdl-21826991

ABSTRACT

OBJECTIVE: To investigate the effects of dimethyloxalyl glycine on hypoxic-ischemic brain damage in newborn rats. METHODS: Forty eight postnatal day 10 SD rats were divided into 3 groups, including sham surgery group, hypoxic-ischemic group and DMOG treated group. The brain tissues were collected at 4, 8, 24 and 72 hours after the hypoxic-ischemic treatment. The expressions of hypoxia inducible factor-1alpha (HIF-1alfa) protein and anti apoptoticprotein cleaved caspase 3 (CC3) were detected by immunohistochemistry. The apoptotic cells were detected by TUNEL staining. RESULTS: The expression level of HIF-1alpha was significantly higher in DMOG treated group than in hypoxic-ischemic group. While the expression level of CC3 was lower and the number of tunel positive cells was fewer in DMOG treated group than that in hypoxic-ischemic group. CONCLUSION: Dimethyloxalyl glycine may play a neuro-protective role in hypoxic-ischemic brain damage in newborn rats by stabilizing HIF-1alpha.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Animals, Newborn , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
16.
Shock ; 36(3): 295-302, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21844787

ABSTRACT

Localized tissue hypoxia is a feature of infection and inflammation, resulting in the upregulation of the transcription factors hypoxia-inducible factor 1α and nuclear factor κB (NF-κB) via inhibition of oxygen sensing hydroxylase enzymes. Previous studies have demonstrated a beneficial role for the hydroxylase inhibitor dimethyloxallyl glycine (DMOG) in inflammatory conditions, including experimental colitis, by regulating the activity of hypoxia-inducible factor 1 and NF-κB. We have demonstrated in vivo that pretreatment with DMOG attenuates systemic LPS-induced activation of the NF-κB pathway. Furthermore, mice treated with DMOG had significantly increased survival in LPS-induced shock. Conversely, in models of polymicrobial sepsis, DMOG exacerbates disease severity. Dimethyloxallyl glycine treatment of mice promotes M2 polarization in macrophages within the peritoneal cavity, resulting in the downregulation of proinflammatory cytokines such as TNF-α. In addition, in vivo DMOG treatment upregulates IL-10 expression, specifically in the peritoneal B1 cell population. This study demonstrates cell type-specific roles for hydroxylase inhibition in vivo and provides insight into the mechanism underlying the protection conveyed by DMOG in models of endotoxic shock.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Macrophages/drug effects , Macrophages/metabolism , Shock, Septic/drug therapy , Animals , Flow Cytometry , Immunoblotting , Interleukin-10/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mixed Function Oxygenases/antagonists & inhibitors , NF-kappa B/metabolism , Polymerase Chain Reaction , Receptors, Interleukin-10/blood , Sepsis/drug therapy , Shock, Septic/chemically induced
17.
Lab Invest ; 91(12): 1684-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21876537

ABSTRACT

Dimethyloxalylglycine (DMOG) is an inhibitor of prolyl-4-hydroxylase domain enzymes. Its potential value and mechanism of actions in preventing/treating gastrointestinal injury are, however, poorly understood. We, therefore, examined the effect of DMOG on influencing gut injury and repair using a variety of in vitro and in vivo models. We performed in vitro studies utilising pro-migratory (wounded monolayer) and proliferation (using DNA quantitation) assays of human stomach (AGS) and colonic (HT29) carcinoma cells. Time course studies examined changes in hypoxia-inducible factor (HIF) and vascular endothelial growth factor (VEGF) levels, a growth factor known to be regulated via HIF. In vivo studies utilised a rat gastric (indomethacin, 20 mg/kg and 3 h restraint) damage model. DMOG stimulated migration in a dose-dependent manner, increasing migration twofold when added at 25µM (P<0.01). Additive effects were seen when DMOG was added to cells in hypoxic conditions. DMOG stimulated proliferation dose dependently, increasing proliferation threefold when added at 70 µM (P<0.01). DMOG caused upregulation of both HIF and VEGF within 4 h of administration. Addition of VEGF neutralising antibody truncated migratory and proliferative activity of DMOG by about 70%. Both oral and subcutaneous administration of DMOG decreased gastric injury without influencing intragastric pH (50% reduction in injury when 1 ml gavaged at 0.57 mM, P < 0.01). Indomethacin reduced tissue HIF and VEGF levels but this was prevented if DMOG was present. In conclusion, DMOG stimulates the early phases of gut repair and VEGF-dependent processes appear relevant. Non-peptide factors such as this may be useful to stabilise or repair gut mucosa.


Subject(s)
Amino Acids, Dicarboxylic/therapeutic use , Gastrointestinal Diseases/drug therapy , Hypoxia-Inducible Factor 1/metabolism , Regeneration/drug effects , Vascular Endothelial Growth Factor A/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , HT29 Cells , Humans , Male , Rats , Rats, Sprague-Dawley
18.
J Immunol ; 186(7): 4367-74, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357264

ABSTRACT

Intestinal ischemia/reperfusion injury (IR) is characterized by intermittent loss of perfusion to the gut, resulting in dramatic increases in morbidity and mortality. Based on previous studies indicating an anti-inflammatory role for hypoxia-inducible factor (HIF)-1-elicited enhancement of extracellular adenosine production via ecto-5'-nucleotidase (CD73) and signaling through the A2B adenosine receptor (A2BAR), we targeted HIF-1 during IR using pharmacological or genetic approaches. Initial studies with pharmacological HIF activation indicated attenuation of intestinal injury with dimethyloxallyl glycine (DMOG) treatment during murine IR. Although DMOG treatment was associated with induction of CD73 transcript and protein, DMOG protection was abolished in cd73(-/-) mice. Similarly, DMOG treatment enhanced A2BAR transcript and protein levels, whereas DMOG protection was abolished in A2BAR(-/-) mice. Finally, studies of mice with conditional HIF-1α deletion in intestinal epithelia or pharmacological inhibition of HIF-1 with 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin revealed enhanced tissue injury during IR. These studies indicated a tissue-protective role of HIF-dependent enhancement of intestinal adenosine generation and signaling during intestinal IR.


Subject(s)
5'-Nucleotidase/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , Receptor, Adenosine A2B/physiology , Reperfusion Injury/prevention & control , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Amino Acids, Dicarboxylic/metabolism , Amino Acids, Dicarboxylic/therapeutic use , Animals , Colitis/enzymology , Colitis/physiopathology , Colitis/prevention & control , Glycine/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/biosynthesis , Reperfusion Injury/enzymology , Reperfusion Injury/physiopathology , Signal Transduction/drug effects , Signal Transduction/immunology
19.
Nat Rev Drug Discov ; 8(2): 139-52, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19165233

ABSTRACT

Cells in the human body need oxygen to function and survive, and severe deprivation of oxygen, as occurs in ischaemic heart disease and stroke, is a major cause of mortality. Nevertheless, other organisms, such as the fossorial mole rat or diving seals, have acquired the ability to survive in conditions of limited oxygen supply. Hypoxia tolerance also allows the heart to survive chronic oxygen shortage, and ischaemic preconditioning protects tissues against lethal hypoxia. The recent discovery of a new family of oxygen sensors--including prolyl hydroxylase domain-containing proteins 1-3 (PHD1-3)--has yielded exciting novel insights into how cells sense oxygen and keep oxygen supply and consumption in balance. Advances in understanding of the role of these oxygen sensors in hypoxia tolerance, ischaemic preconditioning and inflammation are creating new opportunities for pharmacological interventions for ischaemic and inflammatory diseases.


Subject(s)
Hypoxia-Inducible Factor 1/drug effects , Hypoxia/prevention & control , Inflammation/drug therapy , Ischemia/drug therapy , Ischemic Preconditioning/methods , Oxygen Consumption/drug effects , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Amino Acids, Dicarboxylic/pharmacology , Amino Acids, Dicarboxylic/therapeutic use , Angiogenesis Inducing Agents/therapeutic use , Animals , Humans , Hypoxia/metabolism , Inflammation/metabolism , Ischemia/metabolism , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Oxygen Consumption/physiology
20.
Neurobiol Dis ; 31(3): 433-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18602008

ABSTRACT

Hypoxia-inducible factor-1alpha (HIF-1alpha) has been considered as a regulator of both prosurvival and prodeath pathways in the nervous system. The present study was designed to elucidate the role of HIF-1alpha in neonatal hypoxic-ischemic (HI) brain injury. Rice-Vannucci model of neonatal hypoxic-ischemic brain injury was used in seven-day-old rats, by subjecting unilateral carotid artery ligation followed by 2 h of hypoxia (8% O2 at 37 degrees C). HIF-1alpha activity was inhibited by 2-methoxyestradiol (2ME2) and enhanced by dimethyloxalylglycine (DMOG). Results showed that 2ME2 exhibited dose-dependent neuroprotection by decreasing infarct volume and reducing brain edema at 48 h post HI. The neuroprotection was lost when 2ME2 was administered 3 h post HI. HIF-1alpha upregulation by DMOG increased the permeability of the BBB and brain edema compared with HI group. 2ME2 attenuated the increase in HIF-1alpha and VEGF 24 h after HI. 2ME2 also had a long-term effect of protecting against the loss of brain tissue. The study showed that the early inhibition of HIF-1alpha acutely after injury provided neuroprotection after neonatal hypoxia-ischemia which was associated with preservation of BBB integrity, attenuation of brain edema, and neuronal death.


Subject(s)
Brain/drug effects , Brain/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , 2-Methoxyestradiol , Amino Acids, Dicarboxylic/pharmacology , Amino Acids, Dicarboxylic/therapeutic use , Animals , Animals, Newborn , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/physiopathology , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Edema/physiopathology , Brain Infarction/drug therapy , Brain Infarction/metabolism , Brain Infarction/physiopathology , Cytoprotection/drug effects , Cytoprotection/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estradiol/therapeutic use , Female , Hypoxia-Ischemia, Brain/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Tubulin Modulators/pharmacology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...