Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 317(3): F695-F704, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31215802

ABSTRACT

Heme oxygenase (HO) activity is exhibited by inducible (HO-1) and constitutive (HO-2) proteins. HO-1 protects against ischemic and nephrotoxic acute kidney injury (AKI). We have previously demonstrated that HO-2 protects against heme protein-induced AKI. The present study examined whether HO-2 is protective in ischemic AKI. Renal ischemia was imposed on young and aged HO-2+/+ and HO-2-/- mice. On days 1 and 2 after renal ischemia, there were no significant differences in renal function between young male HO-2+/+ and HO-2-/- mice, between young female HO-2+/+ and HO-2-/- mice, or between aged female HO-2+/+ and HO-2-/- mice. However, in aged male mice, HO-2 deficiency worsened renal function on days 1 and 2 after ischemic AKI, and, on day 2 after ischemia, such deficiency augmented upregulation of injury-related genes and worsened histological injury. Renal HO activity was markedly decreased in unstressed aged male HO-2-/- mice and remained so after ischemia, despite exaggerated HO-1 induction in HO-2-/- mice after ischemia. Such exacerbation of deficiency of HO-2 protein and HO activity may reflect phosphorylated STAT3, as activation of this proinflammatory transcription factor was accentuated early after ischemia in aged male HO-2-/- mice. This exacerbation may not reflect impaired induction of nephroprotectant genes, since the induction of HO-1, sirtuin 1, and ß-catenin was accentuated in aged male HO-2-/- mice after ischemia. We conclude that aged male mice are hypersensitive to ischemic AKI and that HO-2 mitigates such sensitivity. We speculate that this protective effect of HO-2 may be mediated, at least in part, by suppression of phosphorylated STAT3-dependent signaling.


Subject(s)
Acute Kidney Injury/prevention & control , Heme Oxygenase (Decyclizing)/metabolism , Kidney/enzymology , Reperfusion Injury/prevention & control , Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Age Factors , Animals , Disease Models, Animal , Female , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Kidney/pathology , Kidney/physiopathology , Male , Mice, Knockout , Phosphorylation , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , STAT3 Transcription Factor/metabolism , Sex Factors , Signal Transduction
2.
J Neuroinflammation ; 13: 26, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26831741

ABSTRACT

BACKGROUND: Following intracerebral hemorrhage (ICH), red blood cells release massive amounts of toxic heme that causes local brain injury. Hemopexin (Hpx) has the highest binding affinity to heme and participates in its transport, while heme oxygenase 2 (HO2) is the rate-limiting enzyme for the degradation of heme. Microglia are the resident macrophages in the brain; however, the significance and role of HO2 and Hpx on microglial clearance of the toxic heme (iron-protoporphyrin IX) after ICH still remain understudied. Accordingly, we postulated that global deletion of constitutive HO2 or Hpx would lead to worsening of ICH outcomes. METHODS: Intracerebral injection of stroma-free hemoglobin (SFHb) was used in our study to induce ICH. Hpx knockout (Hpx(-/-)) or HO2 knockout (HO2(-/-)) mice were injected with 10 µL of SFHb in the striatum. After injection, behavioral/functional tests were performed, along with anatomical analyses. Iron deposition and neuronal degeneration were depicted by Perls' and Fluoro-Jade B staining, respectively. Immunohistochemistry with anti-ionized calcium-binding adapter protein 1 (Iba1) was used to estimate activated microglial cells around the injured site. RESULTS: This study shows that deleting Hpx or HO2 aggravated SFHb-induced brain injury. Compared to wild-type littermates, larger lesion volumes were observed in Hpx(-/-) and HO2(-/-) mice, which also bear more degenerating neurons in the peri-lesion area 24 h postinjection. Fewer Iba1-positive microglial cells were detected at the peri-lesion area in Hpx(-/-) and HO2(-/-) mice, interestingly, which is associated with markedly increased iron-positive microglial cells. Moreover, the Iba1-positive microglial cells increased from 24 to 72 h postinjection and were accompanied with improved neurologic deficits in Hpx(-/-) and HO2(-/-) mice. These results suggest that Iba1-positive microglial cells could engulf the extracellular SFHb and provide protective effects after ICH. We then treated cultured primary microglial cells with SFHb at low and high concentrations. The results show that microglial cells actively take up the extracellular SFHb. Of interest, we also found that iron overload in microglia significantly reduces the Iba1 expression level and resultantly inhibits microglial phagocytosis. CONCLUSIONS: This study suggests that microglial cells contribute to hemoglobin-heme clearance after ICH; however, the resultant iron overloads in microglia appear to decrease Iba1 expression and to further inhibit microglial phagocytosis.


Subject(s)
Brain Injuries/etiology , Brain Injuries/genetics , Cerebral Hemorrhage/complications , Heme Oxygenase (Decyclizing)/deficiency , Hemopexin/deficiency , Acyl-CoA Dehydrogenase/metabolism , Animals , Arabidopsis Proteins/metabolism , Cells, Cultured , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/mortality , Disease Models, Animal , Fluoresceins/metabolism , Heme Oxygenase (Decyclizing)/genetics , Hemoglobins/toxicity , Hemopexin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Motor Activity/genetics , Nerve Tissue Proteins/metabolism , Neurologic Examination , Phagocytosis/drug effects , Phagocytosis/genetics , Time Factors
3.
FASEB J ; 29(1): 105-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25342128

ABSTRACT

Heme oxygenase (HO)-2 deficiency impairs wound healing and exacerbates inflammation following injury. We examine the impact of HO-2 deficiency on macrophage function and the contribution of macrophage HO-2 to inflammatory and repair responses to injury. Corneal epithelial debridement was performed in control and macrophage-depleted HO-2(-/-) and wild-type (WT) mice and in bone marrow chimeras. Peritoneal macrophages were collected for determination of phagocytic activity and classically activated macrophage (M1)-alternatively activated macrophage (M2) polarization. Depletion of macrophages delayed corneal healing (13.2%) and increased neutrophil infiltration (54.1%) by day 4 in WT mice, whereas in HO-2(-/-) mice, it did not worsen the already impaired wound healing and exacerbated inflammation. HO-2(-/-) macrophages displayed an altered M1 phenotype with no significant expression of M2 or M2-like activated cells and a 31.3% reduction in phagocytic capacity that was restored by inducing HO-1 activity or supplementing biliverdin. Macrophage depletion had no effect, whereas adoptive transfer of WT bone marrow improved wound healing (34% on day 4) but did not resolve the exaggerated inflammatory response in HO-2(-/-) mice. These findings indicate that HO-2-deficient macrophages are dysfunctional and that macrophage HO-2 is required for proper macrophage function but is insufficient to correct the impaired healing of the HO-2(-/-) cornea, suggesting that corneal epithelial expression of HO-2 is a key to resolution and repair in wound healing.


Subject(s)
Corneal Injuries/physiopathology , Heme Oxygenase (Decyclizing)/deficiency , Macrophages/enzymology , Macrophages/physiology , Wound Healing/physiology , Animals , Bone Marrow Transplantation , Corneal Injuries/pathology , Cytokines/biosynthesis , Epithelium, Corneal/pathology , Epithelium, Corneal/physiopathology , Female , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/pathology , Inflammation/physiopathology , Macrophage Activation , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/physiology , Phagocytosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transplantation Chimera/physiology
4.
Int J Mol Sci ; 15(10): 17974-99, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25299695

ABSTRACT

Mesenchymal stem cell (MSC) administration is a promising adjuvant therapy to treat tissue injury. However, MSC survival after administration is often hampered by oxidative stress at the site of injury. Heme oxygenase (HO) generates the cytoprotective effector molecules biliverdin/bilirubin, carbon monoxide (CO) and iron/ferritin by breaking down heme. Since HO-activity mediates anti-apoptotic, anti-inflammatory, and anti-oxidative effects, we hypothesized that modulation of the HO-system affects MSC survival. Adipose-derived MSCs (ASCs) from wild type (WT) and HO-2 knockout (KO) mice were isolated and characterized with respect to ASC marker expression. In order to analyze potential modulatory effects of the HO-system on ASC survival, WT and HO-2 KO ASCs were pre-treated with HO-activity modulators, or downstream effector molecules biliverdin, bilirubin, and CO before co-exposure of ASCs to a toxic dose of H2O2. Surprisingly, sensitivity to H2O2-mediated cell death was similar in WT and HO-2 KO ASCs. However, pre-induction of HO-1 expression using curcumin increased ASC survival after H2O2 exposure in both WT and HO-2 KO ASCs. Simultaneous inhibition of HO-activity resulted in loss of curcumin-mediated protection. Co-treatment with glutathione precursor N-Acetylcysteine promoted ASC survival. However, co-incubation with HO-effector molecules bilirubin and biliverdin did not rescue from H2O2-mediated cell death, whereas co-exposure to CO-releasing molecules-2 (CORM-2) significantly increased cell survival, independently from HO-2 expression. Summarizing, our results show that curcumin protects via an HO-1 dependent mechanism against H2O2-mediated apoptosis, and likely through the generation of CO. HO-1 pre-induction or administration of CORMs may thus form an attractive strategy to improve MSC therapy.


Subject(s)
Apoptosis/drug effects , Curcumin/pharmacology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/toxicity , Acetylcysteine/pharmacology , Adipose Tissue/cytology , Animals , Antioxidants/pharmacology , Bilirubin/pharmacology , Biliverdine/pharmacology , Cells, Cultured , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organometallic Compounds/pharmacology , RNA, Messenger/metabolism , Up-Regulation/drug effects
5.
J Cell Mol Med ; 18(12): 2488-98, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224969

ABSTRACT

Impaired wound healing can lead to scarring, and aesthetical and functional problems. The cytoprotective haem oxygenase (HO) enzymes degrade haem into iron, biliverdin and carbon monoxide. HO-1 deficient mice suffer from chronic inflammatory stress and delayed cutaneous wound healing, while corneal wound healing in HO-2 deficient mice is impaired with exorbitant inflammation and absence of HO-1 expression. This study addresses the role of HO-2 in cutaneous excisional wound healing using HO-2 knockout (KO) mice. Here, we show that HO-2 deficiency also delays cutaneous wound closure compared to WT controls. In addition, we detected reduced collagen deposition and vessel density in the wounds of HO-2 KO mice compared to WT controls. Surprisingly, wound closure in HO-2 KO mice was accompanied by an inflammatory response comparable to WT mice. HO-1 induction in HO-2 deficient skin was also similar to WT controls and may explain this protection against exaggerated cutaneous inflammation but not the delayed wound closure. Proliferation and myofibroblast differentiation were similar in both two genotypes. Next, we screened for candidate genes to explain the observed delayed wound closure, and detected delayed gene and protein expression profiles of the chemokine (C-X-C) ligand-11 (CXCL-11) in wounds of HO-2 KO mice. Abnormal regulation of CXCL-11 has been linked to delayed wound healing and disturbed angiogenesis. However, whether aberrant CXCL-11 expression in HO-2 KO mice is caused by or is causing delayed wound healing needs to be further investigated.


Subject(s)
Gene Expression Regulation, Enzymologic , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/genetics , Wound Healing/genetics , Actins/genetics , Actins/metabolism , Animals , Blood Vessels/metabolism , Blotting, Western , Cell Proliferation/genetics , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Collagen/metabolism , Cyclooxygenase 2/metabolism , Gene Expression Profiling , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase-1/metabolism , Immunohistochemistry , Inflammation Mediators/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Skin/injuries , Skin/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
6.
J Am Soc Hypertens ; 7(5): 328-35, 2013.
Article in English | MEDLINE | ID: mdl-23721883

ABSTRACT

BACKGROUND: Heme oxygenase-2 (HO-2) is the main isoform responsible for the breakdown of heme and release of carbon monoxide in the vasculature. Vascular-derived carbon monoxide protects against excessive vasoconstriction due to agents such as angiotensin II (Ang II) and in states of deficiency of nitric oxide. The current study was designed to determine the role of HO-2 in the development of renovascular hypertension using HO-2 knockout mice. METHODS: Polyurethane cuffs were placed around the left renal artery of male and female HO-2 wild-type (WT), heterozygous (HET), and knockout (KO) mice between 16 and 24 weeks of age to induce renovascular hypertension. After 3 weeks, blood pressure was measured for 5 days, after which time both clipped and unclipped kidneys were harvested. RESULTS: No differences were observed in the blood pressure of sham mice between the different genotypes of both sexes. Cuffing of the left renal artery resulted in a significant increase in blood pressure in all genotypes of both sexes. In male mice, the increase in blood pressure was significantly greater in HET and KO mice as compared to WT mice (P < .05). This effect was not observed in female mice. Renovascular hypertension resulted in a significant increase (P < .05) in cardiac hypertrophy in male mice, which was not different between the genotypes. In female mice, HET and KO mice exhibited significantly greater (P < .05) cardiac hypertrophy as compared with WT mice. CONCLUSION: These results demonstrate a sex-specific effect of HO-2 deficiency on the development of renovascular hypertension and its effects on the heart in response to the increase in blood pressure.


Subject(s)
Blood Pressure/genetics , Heme Oxygenase (Decyclizing)/genetics , Hypertension, Renal/genetics , Hypertension, Renal/physiopathology , Sex Characteristics , Angiotensin II/blood , Animals , Bilirubin/blood , Carbon Monoxide/blood , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/immunology , Female , Heme Oxygenase (Decyclizing)/deficiency , Heterozygote , Hypertension, Renal/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Renin/blood , Renin-Angiotensin System/genetics , Renin-Angiotensin System/immunology , Vasoconstriction/physiology
7.
J Neurosci Methods ; 216(2): 128-36, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23583700

ABSTRACT

Neuronal loss in tissue surrounding an intracerebral hemorrhage (ICH) is usually quantified by labor-intensive histological methods that are subject to bias. Fluorescent protein expression has been successfully used as a marker of cell viability in vitro and in retinal studies in vivo, but not in any ICH model to date. The potential of this approach was investigated using transgenic mice that constitutively express the red fluorescent protein variant dTomato in central neurons under the control of the Thy1 promoter. Breeding and growth of these mice were similar to their wild-type counterparts; behavioral phenotyping by digital analysis of home cage video recordings detected no differences. Bright fluorescence was evident in fresh brain samples with minimal background fluorescence, and was reduced in tissue surrounding the hematoma. In order to assess fluorescence loss as an injury marker in a planned study, these mice were crossed with heme oxygenase (HO)-2 knockouts and wild-type controls; striatal hemorrhage was induced by stereotactic injection of collagenase. Fluorescence in hemorrhagic striata was reduced to 86.4±3.9%, 62.2±5.1%, and 58.3±3.0% of contra-lateral on days 1, 4 and 8, respectively, and correlated closely with reduction in striatal cell viability as quantified by MTT assay. HO-2 knockout and wild-type values did not differ significantly. Similar results were observed with stereological cell counts of striatal neurons identified by NeuN immunoreactivity. These results suggest that loss of constitutive dTomato fluorescence is an accurate and efficient marker of neuronal loss in tissue surrounding a striatal hematoma.


Subject(s)
Cerebral Hemorrhage/pathology , Luminescent Agents , Luminescent Proteins , Neurons/pathology , Animals , Cell Survival , Disease Models, Animal , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Time Factors , Transgenes , Red Fluorescent Protein
8.
Am J Physiol Renal Physiol ; 304(3): F317-25, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23195679

ABSTRACT

Age increases the risk for ischemic acute kidney injury (AKI). We questioned whether a similar age-dependent injury occurs following exposure to hemoglobin, a known nephrotoxin. Old mice (~16 mo old), but not young mice (~6 mo old), when administered hemoglobin, exhibited marked elevation in blood urea nitrogen (BUN) and serum creatinine, and acute tubular necrosis with prominent tubular cast formation. The aged kidney exhibited induction of heme oxygenase-1 (HO-1) and other genes/proteins that may protect against heme-mediated renal injury, including ferritin, ferroportin, haptoglobin, and hemopexin. Old mice did not evince induction of HO-2 mRNA by hemoglobin, whereas a modest induction of HO-2 mRNA was observed in young mice. To determine the functional significance of HO-2 in heme protein-induced AKI, we administered hemoglobin to relatively young HO-2(+/+) and HO-2(-/-) mice: HO-2(-/-) mice, compared with HO-2(+/+) mice, exhibited greater renal dysfunction and histologic injury when administered hemoglobin. In addition to failing to elicit a protective system such as HO-2 in response to hemoglobin, old mice exhibited an exaggerated maladaptive response typified by markedly greater induction of the nephrotoxic cytokine IL-6 (130-fold increase vs. 10-fold increase in mRNA in young mice). We conclude that aged mice, unlike relatively younger mice, are exquisitely sensitive to the nephrotoxicity of hemoglobin, an effect attended by a failure to induce HO-2 mRNA and a fulminant upregulation of IL-6. Age thus markedly augments the sensitivity of the kidney to heme proteins, and HO-2 confers resistance to such insults.


Subject(s)
Acute Kidney Injury/chemically induced , Aging/physiology , Hemeproteins/adverse effects , Hemoglobins/adverse effects , Kidney Tubular Necrosis, Acute/chemically induced , Kidney/physiopathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Animals , Blood Urea Nitrogen , Creatinine/blood , Female , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Hemeproteins/metabolism , Hemoglobins/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Kidney Tubular Necrosis, Acute/metabolism , Kidney Tubular Necrosis, Acute/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , RNA, Messenger/metabolism
9.
Tohoku J Exp Med ; 228(1): 27-41, 2012 09.
Article in English | MEDLINE | ID: mdl-22892400

ABSTRACT

Heme is an essential requirement for cell survival. Heme oxygenase (HO) is the rate-limiting enzyme in heme catabolism and consists of two isozymes, HO-1 and HO-2. To identify the protein that regulates the expression or function of HO-1 or HO-2, we searched for proteins that interact with both isozymes, using protein microarrays. We thus identified 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4) that synthesizes or degrades fructose-2,6-bisphosphate, a key activator of glycolysis, depending on cellular microenvironments. Importantly, HO-2 and PFKFB4 are predominantly expressed in haploid spermatids. Here, we show a drastic reduction in expression levels of PFKFB4 mRNA and protein and HO-2 mRNA in HepG2 human hepatoma cells in responses to glucose deprivation (≤ 2.5 mM), which occurred concurrently with remarkable induction of HO-1 mRNA and protein. Knockdown of HO-2 expression in HepG2 cells, using small interfering RNA, caused PFKFB4 mRNA levels to decrease with a concurrent increase in HO-1 expression. Thus, in HepG2 cells, HO-1 expression was increased, when expression levels of HO-2 and PFKFB4 mRNAs were decreased. Conversely, overexpression of HO-2 in HepG2 cells caused the level of co-expressed PFKFB4 protein to increase. These results suggest a potential regulatory role for HO-2 in ensuring PFKFB4 expression. Moreover, in D407 human retinal pigment epithelial cells, glucose deprivation decreased the expression levels of PFKFB4, HO-1, and HO-2 mRNAs. Thus, glucose deprivation consistently down-regulated the expression of PFKFB4 and HO-2 mRNAs in both HepG2 cells and RPE cells. We therefore postulate that PFKFB4 and HO-2 are expressed in a coordinated manner to maintain glucose homeostasis.


Subject(s)
Gene Expression Regulation, Enzymologic , Glycolysis/genetics , Heme Oxygenase (Decyclizing)/genetics , Heme/metabolism , Phosphofructokinase-2/genetics , Animals , Down-Regulation/genetics , Epithelial Cells/enzymology , Gene Knockdown Techniques , Glucose/deficiency , HeLa Cells , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Models, Biological , Organ Specificity , Phosphofructokinase-2/metabolism , Protein Array Analysis , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Testis/enzymology
10.
PLoS One ; 6(6): e21180, 2011.
Article in English | MEDLINE | ID: mdl-21695050

ABSTRACT

Our studies demonstrated that Heme oxygenase (HO), in particular, the constitutive HO-2, is critical for a self-resolving inflammatory and repair response in the cornea. Epithelial injury in HO-2 null mice leads to impaired wound closure and chronic inflammation in the cornea. This study was undertaken to examine the possible relationship between HO-2 and the recruitment of neutrophils following a corneal surface injury in wild type (WT) and HO-2 knockout (HO-2(-/-)) mice treated with Gr-1 monoclonal antibody to deplete peripheral neutrophils. Epithelial injury was performed by removing the entire corneal epithelium. Infiltration of inflammatory cell into the cornea in response to injury was higher in HO-2(-/-) than in WT. However, the rate of corneal wound closure following neutrophil depletion was markedly inhibited in both WT and HO-2(-/-) mice by 60% and 85%, respectively. Neutropenia induced HO-1 expression in WT but not in HO-2(-/-) mice. Moreover, endothelial cells lacking HO-2 expressed higher levels of the Midkine and VE-cadherin and displayed strong adhesion to neutrophils suggesting that perturbation in endothelial cell function caused by HO-2 depletion underlies the increased infiltration of neutrophils into the HO-2(-/-) cornea. Moreover, the fact that neutropenia worsened epithelial healing of the injured cornea in both WT and HO-2(-/-) mice suggest that cells other than neutrophils contribute to the exaggerated inflammation and impaired wound healing seen in the HO-2 null cornea.


Subject(s)
Cornea/immunology , Corneal Injuries , Gene Deletion , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Neutrophils/immunology , Wound Healing/immunology , Animals , Cell Adhesion/immunology , Cell Count , Cornea/enzymology , Cornea/metabolism , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic/immunology , Heme Oxygenase-1/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Neutrophil Infiltration/genetics , Neutrophils/cytology , Neutrophils/enzymology , Neutrophils/metabolism , Wound Healing/genetics
11.
J Cell Physiol ; 226(7): 1732-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506105

ABSTRACT

Heme oxygenase (HO) represents an intrinsic cytoprotective system based on its anti-oxidative and anti-inflammatory properties mediated via its products biliverdin/bilirubin and carbon monoxide (CO). We showed that deletion of HO-2 results in impaired corneal wound healing with associated chronic inflammatory complications. This study was undertaken to examine the role of HO activity and the contribution of HO-1 and HO-2 to corneal wound healing in an in vitro epithelial scratch injury model. A scratch wound model was established using human corneal epithelial (HCE) cells. These cells expressed both HO-1 and HO-2 proteins. Injury elicited a rapid and transient increase in HO-1 and HO activity; HO-2 expression was unchanged. Treatment with biliverdin or CORM-A1, a CO donor, accelerated wound closure by 10% at 24 h. Inhibition of HO activity impaired wound closure by more than 50%. However, addition of biliverdin or CORM-A1 reversed the effect of HO inhibition on wound healing. Moreover, knockdown of HO-2 expression, but not HO-1, significantly impaired wound healing. These results indicate that HO activity is required for corneal epithelial cell migration. Inhibition of HO activity impairs wound healing while amplification of its activity restores and accelerates healing. Importantly, HO-2, which is highly expressed in the corneal epithelium, appears to be critical for the wound healing process in the cornea. The mechanisms by which it contributes to cell migration in response to injury may reside in the cytoprotective properties of CO and biliverdin.


Subject(s)
Epithelium, Corneal/enzymology , Heme Oxygenase (Decyclizing)/deficiency , Wound Healing , Biliverdine/metabolism , Boranes/pharmacology , Carbon Monoxide/metabolism , Carbonates/pharmacology , Cell Movement , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Epithelium, Corneal/drug effects , Epithelium, Corneal/injuries , Epithelium, Corneal/pathology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Humans , RNA Interference , Time Factors , Transfection , Wound Healing/drug effects
12.
J Biochem ; 147(1): 143-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19819905

ABSTRACT

Heme oxygenase (HO) catalyzes oxidative breakdown of heme, and constitutes two isozymes, HO-1 and HO-2. Here, we explored the tissue-specific regulation of expression of HO-1 and HO-2 under hypoxemia. There was no significant change in the overall expression levels of HO-1 and HO-2 mRNAs and proteins in the lung during adaptation of C57BL/6 mice to normobaric hypoxia (10% O(2)). However, immunohistochemical analysis revealed the increased expression of HO-1 and HO-2 proteins after 28 days of normobaric hypoxia in the pulmonary venous myocardium that is the extension of the left atrial myocardium into pulmonary venous walls. Moreover, the expression of HO-2 protein was increased in the sub-endocardial myocardium of ventricles under hypoxia, while HO-1 protein level was increased in the full-thickness walls. Thus, hypoxemia induces expression of both HO-1 and HO-2 proteins in the myocardium. Using C57BL/6 mice lacking HO-2 (HO-2(-/-)), which manifest chronic hypoxemia, we also showed that the HO-1 protein level in the lung was similar between HO-2(-/-) mice and wild-type mice. Unexpectedly, HO-1 protein level was lower by 35% in the HO-2(-/-) mouse liver than the wild-type liver. These results indicate that the expression of HO-1 protein is regulated in a tissue-specific manner under hypoxemia.


Subject(s)
Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase-1/biosynthesis , Hypoxia/enzymology , Myocardium/enzymology , Animals , Heart , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Liver/enzymology , Liver/metabolism , Lung/enzymology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude
13.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1822-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19846746

ABSTRACT

Heme oxygenase (HO) is the enzyme responsible for the breakdown of heme-generating carbon monoxide (CO) and biliverdin in this process. HO-2 is the constitutively expressed isoform in most tissues, such as the kidney and vasculature. CO generated by HO is believed to be an important vasodilator in the renal circulation along with another gas, nitric oxide (NO). To determine the importance of HO-2 in the regulation of blood pressure and renal blood flow (RBF), we treated HO-2 knockout (KO) mice chronically with either ANG II or N(G)-nitroarginine methyl ester (l-NAME). Basal blood pressures were not different between wild-type (WT), heterozygous (HET), or KO mice and averaged 113 +/- 3 vs. 115 +/- 2 vs. 116 +/- 2 mmHg. Similar increases in blood pressure to chronic ANG II as well as l-NAME treatment were observed in all groups with blood pressures increasing an average of 30 mmHg in response to ANG II and 15 mmHg in response to l-NAME. Basal RBFs were not different between the groups averaging 6.0 +/- 0.5 (n = 6) vs. 4.8 +/- 0.6 (n = 10) vs. 5.8 +/- 0.7 (n = 6) ml*min(-1)*g(-1) kidney weight in WT, HET, and KO mice. HO-2 KO and HET mice exhibited an attenuated decrease in RBF in response to acute administration of ANG II, while no differences were observed with l-NAME. Our data indicate that blood pressure and RBF responses to increased ANG II or inhibition of nitric oxide are not significantly enhanced in HO-2 KO mice.


Subject(s)
Blood Pressure , Heme Oxygenase (Decyclizing)/deficiency , Hypertension/enzymology , Kidney/blood supply , Renal Circulation , Administration, Oral , Angiotensin II/administration & dosage , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Disease Models, Animal , Female , Heme Oxygenase (Decyclizing)/genetics , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Infusion Pumps, Implantable , Infusions, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/administration & dosage , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Vascular Resistance , Vasoconstriction
14.
J Pharmacol Exp Ther ; 331(3): 925-32, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19773531

ABSTRACT

In previous studies, we have shown that heme oxygenase (HO)-2 null [HO-2(-/-)] mice exhibit a faulty response to injury; chronic inflammation and massive neovascularization replaced resolution of inflammation and tissue repair. Endothelial cells play an active and essential role in the control of inflammation and the process of angiogenesis. We examined whether HO-2 deletion affects endothelial cell function. Under basal conditions, HO-2(-/-) aortic endothelial cells (mAEC) showed a 3-fold higher expression of vascular endothelial growth factor receptor 1 and a marked angiogenic response compared with wild-type (WT) cells. Compared with WT cells, HO-2(-/-) mAEC showed a 2-fold reduction in HO activity and marked increases in levels of gp91(phox)/NADPH oxidase isoform, superoxide, nuclear factor kappaB activation, and expression of inflammatory cytokines, including interleukin (IL)-1alpha and IL-6. HO-2 deletion transforms endothelial cells from a "normal" to an "activated" phenotype characterized by increases in inflammatory, oxidative, and angiogenic factors. This switch may be the result of reduced HO activity and the associated reduction in the cytoprotective HO products, carbon monoxide and biliverdin/bilirubin, because addition of biliverdin to HO-2(-/-) cells attenuated angiogenesis and reduced superoxide production. This transformation underscores the importance of HO-2 in the regulation of endothelial cell homeostasis.


Subject(s)
Endothelial Cells/enzymology , Endothelium, Vascular/enzymology , Gene Deletion , Heme Oxygenase (Decyclizing)/deficiency , Inflammation/enzymology , Neovascularization, Pathologic/enzymology , Oxidative Stress , Animals , Blotting, Western , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Microscopy, Fluorescence , NF-kappa B/immunology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oxidative Stress/genetics , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Superoxides/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
15.
Neuroscience ; 155(4): 1133-41, 2008 Sep 09.
Article in English | MEDLINE | ID: mdl-18674596

ABSTRACT

Intracerebral hemorrhage (ICH) remains a major medical problem and currently has no effective treatment. Hemorrhaged blood is highly toxic to the brain, and catabolism of the pro-oxidant heme, mainly released from hemoglobin, is critical for the resolution of hematoma after ICH. The degradation of the pro-oxidant heme is controlled by heme oxygenase (HO). We have previously reported a neuroprotective role for HO2 in early brain injury after ICH; however, in vivo data that specifically address the role of HO2 in brain edema and neuroinflammation after ICH are absent. Here, we tested the hypothesis that HO2 deletion would exacerbate ICH-induced brain edema, neuroinflammation, and oxidative damage. We subjected wild-type (WT) and HO2 knockout ((-/-)) mice to the collagenase-induced ICH model. Interestingly, HO2(-/-) mice had enhanced brain swelling and neuronal death, although HO2 deletion did not increase collagenase-induced bleeding; the exacerbation of brain injury in HO2(-/-) mice was also associated with increases in neutrophil infiltration, microglial/macrophage and astrocyte activation, DNA damage, peroxynitrite production, and cytochrome c immunoreactivity. In addition, we found that hemispheric enlargement was more sensitive than brain water content in the detection of subtle changes in brain edema formation in this model. Combined, these novel findings extend our previous observations and demonstrate that HO2 deficiency increases brain swelling, neuroinflammation, and oxidative damage. The results provide additional evidence that HO2 plays a critical protective role against ICH-induced early brain injury.


Subject(s)
Brain Edema/etiology , Brain Edema/genetics , Cerebral Hemorrhage/complications , Encephalitis/etiology , Encephalitis/genetics , Heme Oxygenase (Decyclizing)/deficiency , Analysis of Variance , Animals , Calcium-Binding Proteins/metabolism , Cytochromes c/metabolism , Disease Models, Animal , Fluoresceins , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-3/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Nerve Degeneration/etiology , Organic Chemicals , Recombinant Fusion Proteins/metabolism , Recombinant Proteins , Spectrophotometry/methods , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
Exp Eye Res ; 87(3): 268-78, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18602389

ABSTRACT

Heme oxygenase (HO-1 and HO-2) represents an intrinsic cytoprotective and anti-inflammatory system based on its ability to modulate leukocyte migration and to inhibit expression of inflammatory cytokines and proteins. HO-2 deletion leads to unresolved corneal inflammation and chronic inflammatory complications including ulceration, perforation and neovascularization. We examined the consequences of HO-2 deletion on hemangiogenesis and lymphangiogenesis in the model of suture-induced inflammatory neovascularization. An 8.0 silk suture was placed at the corneal apex of wild type and HO-2 null mice. Neovascularization was assessed by vital microscopy and quantified by image analysis. Hemangiogenesis and lymphangiogenesis were determined by immunofluorescence staining using anti-CD31 and anti-LYVE-1 antibodies, respectively. Inflammation was quantified by histology and myeloperoxidase activity. The levels of HO-1 expression and inflammatory cytokines were determined by real time PCR and ELISA, respectively. Corneal sutures produced a consistent inflammatory response and a time-dependent neovascularization. The response in HO-2 null mice was associated with a greater increase compared to the wild type in the number of leukocytes (827,600+/-129,000 vs. 294,500+/-57,510; p<0.05), neovessels measured by vital microscopy (21.91+/-1.05 vs. 12.77+/-1.55 mm; p<0.001) 4 days after suture placement. Hemangiogenesis but not lymphangiogenesis was more pronounced in HO-2 null mice compared to wild type mice. Induction of HO-1 in sutured corneas was greatly attenuated in HO-2 null corneas and treatment with biliverdin diminished the exaggerated inflammatory and neovascular response in HO-2 null mice. The demonstration that the inflammatory responses, including expression of proinflammatory proteins, inflammatory cell influx and hemangiogenesis are exaggerated in HO-2 knockout mice strongly supports the notion that the HO system is critical for controlling the inflammatory and neovascular response in the cornea. Hence, pharmacological amplification of this system may constitute a novel therapeutic strategy for the treatment of corneal disorders associated with excessive inflammation and neovascularization.


Subject(s)
Biliverdine/therapeutic use , Corneal Neovascularization/prevention & control , Keratitis/prevention & control , Animals , Cornea/enzymology , Corneal Neovascularization/enzymology , Corneal Neovascularization/pathology , Disease Progression , Drug Evaluation, Preclinical , Female , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1/metabolism , Keratitis/enzymology , Keratitis/pathology , Male , Mice , Mice, Knockout , Peroxidase/metabolism
17.
Am J Physiol Regul Integr Comp Physiol ; 295(2): R498-504, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495834

ABSTRACT

Carbon monoxide derived from heme oxygenase (HO) may participate in cerebrovascular regulation under specific circumstances. Previous work has shown that HO contributes to feline pial arteriolar dilation to acetylcholine after transfusion of a cell-free polymeric hemoglobin oxygen carrier. The role of constitutive HO2 in the pial arteriolar dilatory response to acetylcholine was determined by using 1) HO2-null mice (HO2-/-), 2) the HO inhibitor tin protoporphyrin IX (SnPPIX), and 3) 4,5,6,7-tetrabromobenzotriazole (TBB), an inhibitor of casein kinase-2 (CK2)-dependent phosphorylation of HO2. In anesthetized mice, superfusion of a cranial window with SnPPIX decreased arteriolar dilation produced by 10 microM acetylcholine by 51%. After partial polymeric hemoglobin exchange transfusion, the acetylcholine response was normal but was reduced 72% by SnPPIX and 95% by TBB. In HO2-/- mice, the acetylcholine response was modestly reduced by 14% compared with control mice and was unaffected by SnPPIX. After hemoglobin transfusion in HO2-/- mice, acetylcholine responses were also unaffected by SnPPIX and TBB. In contrast, nitric oxide synthase inhibition completely blocked the acetylcholine responses in hemoglobin-transfused HO2-/- mice. We conclude 1) that HO2 activity partially contributes to acetylcholine-induced pial arteriolar dilation in mice, 2) that this contribution is augmented in the presence of a plasma-based hemoglobin polymer and appears to depend on a CK2 kinase mechanism, 3) that nitric oxide synthase activity rather than HO1 activity contributes to the acetylcholine reactivity in HO2-/- mice, and 4) that plasma-based polymeric hemoglobin does not scavenge all of the nitric oxide generated by cerebrovascular acetylcholine stimulation.


Subject(s)
Acetylcholine/pharmacology , Blood Component Transfusion , Blood Substitutes/administration & dosage , Cerebrovascular Circulation/drug effects , Cerebrum/blood supply , Heme Oxygenase (Decyclizing)/metabolism , Hemoglobins/administration & dosage , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Arterioles/drug effects , Arterioles/enzymology , Carbon Dioxide/blood , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitroarginine/pharmacology , Protoporphyrins/pharmacology , Triazoles/pharmacology
18.
Am J Physiol Gastrointest Liver Physiol ; 293(2): G438-45, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17510199

ABSTRACT

The aims of this study were to quantify the change in resting membrane potential (RMP) across the thickness of the circular muscle layer in the mouse and human small intestine and to determine whether the gradient in RMP is dependent on the endogenous production of carbon monoxide (CO). Conventional sharp glass microelectrodes were used to record the RMPs of circular smooth muscle cells at different depths in the human small intestine and in wild-type, HO2-KO, and W/W(V) mutant mouse small intestine. In the wild-type mouse and human intestine, the RMP of circular smooth muscle cells near the myenteric plexus was -65.3 +/- 2 mV and -58.4 +/- 2 mV, respectively, and -60.1 +/- 2 mV and -49.1 +/- 1 mV, respectively, in circular smooth muscle cells at the submucosal border. Oxyhemoglobin (20 microM), a trapping agent for CO, and chromium mesoporphyrin IX, an inhibitor of heme oxygenase, abolished the transwall gradient. The RMP gradients in mouse and human small intestine were not altered by N(G)-nitro-l-arginine (200 microM). No transwall RMP gradient was found in HO2-KO mice and W/W(V) mutant mice. TTX (1 microM) and 1H-[1,2,4-]oxadiazolo[4,3-a]quinoxalin-1-one (10 microM) had no effect on the RMP gradient. These data suggest that the gradient in RMP across the thickness of the circular muscle layer of mouse and human small intestine is CO dependent.


Subject(s)
Carbon Monoxide/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Intestine, Small/metabolism , Muscle, Smooth/metabolism , Animals , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Humans , In Vitro Techniques , Intestine, Small/drug effects , Intestine, Small/enzymology , Membrane Potentials , Mesoporphyrins/pharmacology , Mice , Mice, Knockout , Mice, Neurologic Mutants , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitroarginine/pharmacology , Oxadiazoles/pharmacology , Oxyhemoglobins/metabolism , Quinoxalines/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Tetrodotoxin/pharmacology
19.
FASEB J ; 21(9): 2257-66, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17384141

ABSTRACT

In the immune-privileged cornea, epithelial wounds heal rapidly with almost no scarring and, unlike in most other tissues, acute inflammation in the absence of infection is beneficial to healing. Molecular mechanisms, which account for this striking property, remain to be clearly defined, but they likely include autacoids that control leukocyte activation. Two prominent enzymes, 12/15-lipoxygenase (LOX), which generates antiinflammatory lipid autacoids, and heme-oxygenase (HO), which generates antioxidants and carbon monoxide, are highly expressed in human and mouse corneas. LXA4, an endogenous 12/15-LOX product, proved to be a potent inhibitor of exacerbated inflammation and significantly increased re-epithelialization in corneal wounds. In vivo deletion of 12/15-LOX correlated with exacerbated inflammation and impaired wound healing in 12/15-LOX(-/-) mice, a phenotype that was rescued by treatment with LXA4. More importantly, 12/15-LOX(-/-) mice demonstrated impaired induction of HO-1 in both acute and exacerbated inflammation. Topical LXA4 restored HO-1 expression in 12/15-LOX(-/-) mice and amplified HO-1 gene expression in human corneal epithelial cells. HO-2(-/-) mice, which fail to induce HO-1, also demonstrated exacerbated inflammation in response to injury, a phenotype that, notably, correlated with a 50% reduction in endogenous LXA4 formation. Collectively, results demonstrate a critical role for LXA4 in inflammatory/reparative responses and provide the first evidence that 12/15-LOX and HO systems function in concert to control inflammation.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Epithelium, Corneal/injuries , Heme Oxygenase (Decyclizing)/physiology , Heme Oxygenase-1/physiology , Keratitis/physiopathology , Lipoxins/physiology , Membrane Proteins/physiology , Multienzyme Complexes/physiology , Wound Healing/physiology , Administration, Topical , Animals , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/genetics , Autacoids/pharmacology , Cells, Cultured , Epithelium, Corneal/cytology , Epithelium, Corneal/metabolism , Eye Injuries/drug therapy , Feedback, Physiological , Female , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/genetics , Humans , Keratitis/chemically induced , Keratitis/drug therapy , Lipopolysaccharides/toxicity , Lipoxins/administration & dosage , Lipoxins/biosynthesis , Lipoxins/pharmacology , Lipoxins/therapeutic use , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Wound Healing/drug effects
20.
Am J Pathol ; 169(5): 1612-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17071585

ABSTRACT

Heme oxygenase (HO) represents an intrinsic anti-inflammatory system based on its ability to regulate leukocyte function and inhibit expression of proinflammatory cytokines. This anti-inflammatory function is linked to the inducible isoform HO-1; the role of the constitutive isoform HO-2 is unknown. The current study was undertaken to investigate the role of HO-2 in the regulation of the acute inflammatory and reparative response by using HO-2-null mice and well-established animal models of epithelial injury and antigen-induced peritonitis. Here we show that in vivo deletion of HO-2 disables execution of the acute inflammatory and reparative response after epithelial injury and leads to an exaggerated inflammatory response in antigen-induced peritonitis. HO-2 deletion was associated with impaired HO-1 induction, indicating that HO-2 is critical for HO-1 expression and that the subsequent failure to up-regulate the HO system may contribute to unresolved inflammation and the development of chronic inflammatory conditions. Indeed, supplementation with the HO bioactive product, biliverdin, rescued the acute inflammatory and reparative response in HO-2-null mice. Thus, HO-2 sets in place a basal tone of anti-inflammatory signals that may be a prerequisite for the ordered execution of an inflammatory and reparative response.


Subject(s)
Heme Oxygenase (Decyclizing)/metabolism , Inflammation/immunology , Wound Healing/physiology , Animals , Biliverdine/pharmacology , Cornea/blood supply , Cornea/cytology , Cornea/drug effects , Cornea/pathology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase (Decyclizing)/deficiency , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , Leukocytes/immunology , Lipid Metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...