Article

Involvement of heme oxygenase-1 (HO-1) in the adaptive protection of human lymphocytes after hyperbaric oxygen (HBO) treatment

Authors:
To read the full-text of this research, you can request a copy directly from the author.

Abstract

Accumulating evidence suggests that HO-1 plays an important role in cellular protection against oxidant-mediated cell injury. Our previous studies on hyperbaric oxygen (HBO; i.e. exposure to pure oxygen under high ambient pressure) indicated clearly increased levels of HO-1 in lymphocytes of volunteers 24 h after HBO treatment (1 h at 1.5 bar). Experiments with the comet assay (alkaline single cell gel electrophoresis) revealed that the same cells were almost completely protected against the induction of DNA damage by a repeated exposure or in vitro treatment with H 2O2 24 h after the first HBO. In order to further investigate the role of HO-1 in HBO-induced adaptive response, we now performed experiments with isolated human lymphocytes exposed to HBO in vitro (2 h at 3 bar). Our results show that also under cell culture conditions, lymphocytes exhibit an adaptive protection similar to that observed in our previous work with healthy human subjects. The timecourse of HO-1 induction proceeds in parallel to the development of an adaptive protection against the induction of oxidative DNA damage. A comparable protection was not seen in V79 cells, indicating a specific difference between the two investigated cell systems. Treatment with the specific HO-1 inhibitor tin-mesoporphyrin IX (SnMP) led to a complete abrogation of HBO-induced adaptive protection in human lymphocytes. Our results indicate a functional involvement of HO-1 in the adaptive protection of human lymphocytes against the induction of oxidative DNA damage. The exact mechanism by which HO-1 contributes to an adaptive response remains to be elucidated.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the author.

... This increased oxygen content is known to cause several cellular effects including the inhibition of interferon-c [12], interleukin-1b and tumor necrosis factor a release [13], a temporary drop in the CD4:CD8 lymphocyte ratio [14], upregulation of the interleukin-2 receptor and downregulation of the ab T-cell receptor [15], induction of DNA damage and compensatory heme oxygenase-1 upregulation [16][17][18][19][20], decrease of lymphocyte proliferation [21] and inhibition of human mammary epithelial proliferation [22]. As an overall effect, at least in rodents, hyperbaric oxygenation appears to be antiinflammatory since it increases the susceptibility to respiratory infections [23,24] and delays allograft rejection [25][26][27]. ...
... Similarly, repeated HBO elevated hippocampal Bcl-2 expression in gerbils [63]. Heme-oxygenase-1 is known to be upregulated in response to HBO in cell cultures [18] as well as in human volunteers [20]. Induction of heme oxygenase-1 protected against HBO-mediated DNA-damage [16][17][18]20]. ...
... Heme-oxygenase-1 is known to be upregulated in response to HBO in cell cultures [18] as well as in human volunteers [20]. Induction of heme oxygenase-1 protected against HBO-mediated DNA-damage [16][17][18]20]. In analogy to the findings regarding heme oxygenase-1 we propose that the upregulation of Bcl-2 may be a protective mechanism of counter regulation in response to proapoptotic stress by hyperbaric oxygen. ...
Article
During therapeutic hyperbaric oxygenation lymphocytes are exposed to high partial pressures of oxygen. This study aimed to analyze the mechanism of apoptosis induction by hyperbaric oxygen. For intervals of 0.5-4 h Jurkat-T-cells were exposed to ambient air or oxygen atmospheres at 1-3 absolute atmospheres. Apoptosis was analyzed by phosphatidylserine externalization, caspase-3 activation and DNA-fragmentation using flow cytometry. Apoptosis was already induced after 30 min of hyperbaric oxygenation (HBO, P < 0.05). The death receptor Fas was downregulated. Inhibition of caspase-9 but not caspase-8 blocked apoptosis induction by HBO. Hyperbaric oxygen caused a loss of mitochondrial membrane potential and caspase-9 induction. The mitochondrial pro-survival protein Bcl-2 was upregulated, and antagonizing Bcl-2 function potentiated apoptosis induction by HBO. In conclusion, a single exposure to hyperbaric oxygenation induces lymphocyte apoptosis by a mitochondrial and not a Fas-related mechanism. Regulation of Fas and Bcl-2 may be regarded as protective measures of the cell in response to hyperbaric oxygen.
... The concept of HBOT-related protection and repair of renal tissues is supported by our novel finding that HBOT upregulates NRF2-dependent antioxidant pathways with in tissues of the obese T2DM mouse. Furthermore, the family of molecular chaperones (HSPs and hemoxygenase-1) which plays critical roles in protein folding and attenuation of oxidative damage in tissues was also activated by HBOT (Rothfuss et al. 2001;Godman et al. 2010). These protective antioxidant genes may be repressed due to tissue hypoxia within or adjacent to the site of tissue damage and inflammation. ...
... The concomitant decrease in vitamin C metabolites, such as ascorbate (0.79, p < 0.05), following HBOT suggests that there is an increase in the detoxification of oxygen radicals or potentially better tubular reabsorption of vitamin C. This observation is consistent with increased oxidative signaling and antioxidant defenses arising from higher tissue oxygen tensions after HBOT exposure (Rothfuss et al. 2001;Bader et al. 2007) (Fig. 1). White background represents p < 0.05, gray is 0.05 < p < 0.10. ...
Article
Diabetic kidney disease (DKD) is the leading cause of end-stage renal failure in the western world. Current treatment of diabetic kidney disease relies on nutritional management and drug therapies to achieve metabolic control. Here, we discuss the potential application of hyperbaric oxygen therapy (HBOT) for the treatment of diabetic kidney disease (DKD), a treatment which requires patients to breathe in 100% oxygen at elevated ambient pressures. HBOT has traditionally been used to diabetic foot ulcers (DFU) refractory to conventional medical treatments. Successful clinic responses seen in the DFU provide the underlying therapeutic rationale for testing HBOT in the setting of DKD. Both the DFU and DKD have microvascular endothelial disease as a common underlying pathologic feature. Supporting evidence for HBOT of DKD comes from previous animal studies and from our preliminary prospective clinical trial reported here. We report urinary metabolomic data obtained from patients undergoing HBOT for DFU, before and after exposure to 6 weeks of HBOT. The preliminary data support the concept that HBOT can reduce biomarkers of renal injury, oxidant stress, and mitochondrial dysfunction in patients receiving HBOT for DFU. Further studies are needed to confirm these initial findings and correlate them with simultaneous measures of renal function. HBOT is a safe and effective treatment for DFU and could also be for individuals with DKD.
... The expression of HO-1 is dependent on the cell type, cellular microenvironment, intensity and duration of stimuli exposure, and is regulated by a panel of redox-sensitive transcription factors, including HIF-1α and NF-κB [69]. In numerous studies and circumstances, HBO 2 treatment has been linked to a protective impact of enhanced HO-1 activity [70][71][72][73][74][75]. Most pertinent to this review is that levels of HO-1 have been shown to increase in a subgroup of NSTI patients with septic shock in response to HBO 2 treatment [24]. ...
Article
Full-text available
The perception of sepsis has shifted over time; however, it remains a leading cause of death worldwide. Sepsis is now recognized as an imbalance in host cellular functions triggered by the invading pathogens, both related to immune cells, endothelial function, glucose and oxygen metabolism, tissue repair and restoration. Many of these key mechanisms in sepsis are also targets of hyperbaric oxygen (HBO2) treatment. HBO2 treatment has been shown to improve survival in clinical studies on patients with necrotizing soft tissue infections as well as experimental sepsis models. High tissue oxygen tension during HBO2 treatment may affect oxidative phosphorylation in mitochondria. Oxygen is converted to energy, and, as a natural byproduct, reactive oxygen species are produced. Reactive oxygen species can act as mediators, and both these and the HBO2-mediated increase in oxygen supply have the potential to influence the cellular processes involved in sepsis. The pathophysiology of sepsis can be explained comprehensively through resistance and tolerance to infection. We argue that HBO2 treatment may protect the host from collateral tissue damage during resistance by reducing neutrophil extracellular traps, inhibiting neutrophil adhesion to vascular endothelium, reducing proinflammatory cytokines, and halting the Warburg effect, while also assisting the host in tolerance to infection by reducing iron-mediated injury and upregulating anti-inflammatory measures. Finally, we show how inflammation and oxygen-sensing pathways are connected on the cellular level in a self-reinforcing and detrimental manner in inflammatory conditions, and with support from a substantial body of studies from the literature, we conclude by demonstrating that HBO2 treatment can intervene to maintain homeostasis.
... In the HOB cells, an expression response to HBO was evident from the significant increase in NQO1 expression 24 h after HBO exposure, and this is in good correlation with our findings on HO-1 expression. Thus, the cellular response to oxidative stress was similar in the expression of HO-1 and NQO1, and appears to function similarly in osteoblasts as in lymphocytes or lung cells [50,54,55]. However, the expression of NQO1 in the SAOS-2 cells was not affected by HBO exposure and remained at very low levels at all time points. ...
Article
Full-text available
(1) Background: Hyperbaric oxygen (HBO) exposure induces oxidative stress that may lead to DNA damage, which has been observed in human peripheral blood lymphocytes or non-human cells. Here, we investigated the impact of hyperbaric conditions on two human osteoblastic cell lines: primary human osteoblasts, HOBs, and the osteogenic tumor cell line SAOS-2. (2) Methods: Cells were exposed to HBO in an experimental hyperbaric chamber (4 ATA, 100% oxygen, 37 °C, and 4 h) or sham-exposed (1 ATA, air, 37 °C, and 4 h). DNA damage was examined before, directly after, and 24 h after exposure with an alkaline comet assay and detection of γH2AX+53BP1 colocalizing double-strand break (DSB) foci and apoptosis. The gene expression of TGFß-1, HO-1, and NQO1, involved in antioxidative functions, was measured with qRT-PCR. (3) Results: The alkaline comet assay showed significantly elevated levels of DNA damage in both cell lines after 4 h of HBO, while the DSB foci were similar to sham. γH2AX analysis indicated a slight increase in apoptosis in both cell lines. The increased expression of HO-1 in HOB and SAOS-2 directly after exposure suggested the induction of an antioxidative response in these cells. Additionally, the expression of TGF-ß1 was negatively affected in HOB cells 4 h after exposure. (4) Conclusions: in summary, this study indicates that osteoblastic cells are sensitive to the DNA-damaging effects of hyperbaric hyperoxia, with the HBO-induced DNA damage consisting largely of single-strand DNA breaks that are rapidly repaired.
... Experimental evidence has shown that HBO 2 therapy protects against mitochondrial dysfunction [35] and improves mitochondrial biogenesis and respiration [36,37], thus supporting its beneficial and therapeutic effects. Furthermore, HBO 2 therapy can reverse CO binding of COX [7,38] and reduce oxidative stress via various mechanisms, including increased hem oxygenase-1 [39,40], the upregulation of antioxidant enzymes [31,41], and the induction of the heat shock protein levels, which protects against oxidative stressinduced damage [42]. ...
Article
Full-text available
Mitochondrial and oxidative stress play critical roles in the pathogenic mechanisms of carbon monoxide (CO)-induced toxicity. This study was designed to evaluate whether the serum levels of specific stress biomarkers might reflect brain injury and act as prognostic markers for the development of neurocognitive sequelae following CO poisoning. We analyzed the data from 51 adult patients admitted with acute CO poisoning and measured the serum level expression of growth differentiation factor 15 (GDF15) and fibroblast growth factor 21 (FGF21), indicators of mitochondrial stress, and 8-Oxo-2′-deoxyguanosine (8-OHdG) and malondialdehyde (MDA), indicators of oxidative stress. Serum was collected upon arrival at the hospital, at 24 h post treatment, and within 7 days of HBO2 therapy. Global Deterioration Scale scores were measured 1 month post incident and used to place the patients in either favorable or poor outcome groups. Initial serum GDF15 and 8-OHdG concentrations were significantly increased in the poor-outcome group and all four biomarkers decreased at 24 h post HBO2 therapy, and were then maintained or further decreased at the 1-week mark. Notably, the degree of change in these biomarkers between baseline and 24 h post HBO2 were significantly larger in the poor-outcome group, reflecting greater CO-associated stress, confirming that post-CO poisoning serum biomarker levels and their response to HBO2 were proportional to the initial stress. We suggest that these biomarkers accurately reflect neuronal toxicity in response to CO poisoning, which is consistent with their activity in other pathologies.
... HBO treatment considerably accelerated bone formation by increasing osteogenic differentiation of those MSCs [13]. Furthermore, HBO had a protective impact on human lymphocytes by preventing DNA damage in the course of oxidant-mediated cell injury [14]. ...
Article
Full-text available
For more than six decades, hyperbaric oxygen (HBO) has been used for a variety of indications involving tissue repair. These indications comprise a wide range of diseases ranging from intoxications to ischemia-reperfusion injury, crush syndrome, central nervous injury, radiation-induced tissue damage, burn injury and chronic wounds. In a systematic review, the molecular mechanisms triggered by HBO described within the last two decades were compiled. They cover a wide range of pathways, including transcription, cell-to-cell contacts, structure, adhesion and transmigration, vascular signaling and response to oxidative stress, apoptosis, autophagy and cell death, as well as inflammatory processes. By analyzing 71 predominantly experimental publications, we established an overview of the current concepts regarding the molecular mechanisms underlying the effects of HBO. We considered both the abovementioned pathways and their role in various applications and indications.
... 22 Although high oxygen levels produced by hyperbaric oxygen are only maintained when the patient is in the hyperbaric oxygen chamber, and for a short time afterward, HBO 2 can also produce various biochemical effects, including (1) inhibition of neutrophils' adhesion and production of proinflammatory cytokines (interleukin-1 (IL-1), IL-6 and TNF-a), (2) upregulation of hypoxia response pathway (HIF-1α, HO-1), (3) changes in host-microbiome metabolism and (4) increased growth factor synthesis and migration. [26][27][28][29][30][31] OBJECTIVES Some studies have demonstrated that HBO 2 can relieve a range of symptoms of patients who suffer from moderateto-severe UC. On the contrary, Pagoldh et al conducted a prospective randomised study and indicated that HBO 2 is ineffective in treating UC. 32 Dulai et al conducted a systematic review of safety and effectiveness of hyperbaric oxygen in treating IBD (including Crohn's disease and UC) in 2014, and they concluded that hyperbaric oxygen is a relatively safe and potentially effective option for IBD treatment. ...
Article
Full-text available
Introduction Ulcerative colitis (UC) is a type of inflammatory bowel disease, and 62% of patients with UC felt that it is difficult for them to live a normal life. Furthermore, some researches have shown that about 15% of patients with UC undergo at least one extreme clinical course in their lifetime, and 10%–30% of patients with UC oblige colectomy. Although many investigations have demonstrated that HBO 2 has a beneficial impact on UC treatment, a systematic review and meta-analysis are unavailable. Therefore, a meta-analysis is essential to assess the efficacy and safety of HBO 2 in treating UC. Methods and analysis A systematic search plan will be performed in the following seven databases with a restriction of time from inception to September 2020 to filter the eligible studies: PubMed, Web of Science, Embase, Cochrane Library, China National Knowledge Infrastructure, Chinese Scientific Journal Database (VIP) and Chinese Biomedical Database WanFang. Other related resources will be also searched. Two independent reviewers will choose eligible researches and extract data. The risk of bias will be evaluated based on Cochrane Collaboration’s Risk of Bias tool and Newcastle-Ottawa Scale. Eventually, a systematic review and meta-analysis will be performed via the Review Manager V.5.3 statistical software and STATA V.14.0 software. Ethics and dissemination This study will not involve the individual patient and any ethical problems since its outcomes are based on published data. Therefore, no ethical review and approval are required. We plan to publish the study in a peer-reviewed journal. PROSPERO registration number CRD42020210244.
... 20 21 Moreover, HBO 2 -mediated alterations of the two antioxidants, heme oxygenase-1 (HO-1) and superoxide dismutase (SOD), have been demonstrated in experimental models of sepsis associated acute lung injury, in sepsis, and as part of a protective mechanism against oxidative DNA damage. [22][23][24][25][26][27] Multiple HBO 2 treatments have been shown to elevate RNS by enhancing production of nitrite+nitrate in selected cohorts, 28 but the systemic effect on RNS remains unresolved. 29 30 Of notable interest is a SODmediated regulatory function of NO expression 31 and an inhibitory function of HO-1 on the NO synthase expression, 32 indicating closely interconnected regulatory mechanisms between RNS and ROS. ...
Article
Full-text available
Necrotizing soft-tissue infection (NSTI) is a rare, severe, and fast-progressing bacterial infection associated with a high risk of developing sepsis or septic shock. Increasing evidence indicates that oxidative stress is crucial in the development and progression of sepsis, but its role in NSTI specifically has not been investigated. Some patients with NSTI receive hyperbaric oxygen (HBO 2 ) treatment as the restoration of oxidative stress balance is considered an important mechanism of action, which HBO 2 facilitates. However, a gap in knowledge exists regarding the effect of HBO 2 treatment on oxidative stress in patients with NSTI. In the present observational study, we aimed to investigate HBO 2 treatment effects on known markers of oxidative stress in patients with NSTI. We measured plasma myeloperoxidase (MPO), superoxide dismutase (SOD), heme oxygenase-1 (HO-1) and nitrite+nitrate in 80 patients with NSTI immediately before and after their first HBO 2 treatment, and on the following day. We found that HBO 2 treatment was associated with a significant increase in MPO and SOD by a median of 3.4 and 8.8 ng/mL, respectively. Moreover, we observed an HBO 2 treatment-associated increase in HO-1 in patients presenting with septic shock (n=39) by a median of 301.3 pg/mL. All markers were significantly higher in patients presenting with septic shock compared to patients without shock, and all markers correlated with disease severity. High baseline SOD was associated with 90-day mortality. In conclusion, HBO 2 treatment was associated with an increase in MPO and SOD in patients with NSTI, and oxidative stress was more pronounced in patients with septic shock.
... HO-1, also called heat shock protein (HSP)32 is the first molecular which is reported to be a mediator for achieved improvement induced by HBO, particularly by increasing the tolerance of the organism against oxidative damage [40]. Rothfuss et al. demonstrated involvement of HO-1 in the adaptive protection of human lymphocytes after hyperbaric oxygen treatment [41]. Our results also showed no statistical difference in HO-1 expression between SHAM and SHR -AKI. ...
Article
Full-text available
: Renal ischemia and reperfusion (I/R) injury is the most common cause of acute kidney injury (AKI). Pathogenesis of postischemic AKI involves hemodynamic changes, oxidative stress, inflammation process, calcium ion overloading, apoptosis and necrosis. Up to date, therapeutic approaches to treat AKI are extremely limited. Thus, the aim of this study was to evaluate the effects of hyperbaric oxygen (HBO) preconditioning on citoprotective enzyme, heme oxygenase-1 (HO-1), pro-apoptotic Bax and anti-apoptotic Bcl-2 proteins expression, in postischemic AKI induced in normotensive Wistar and spontaneously hypertensive rats (SHR). The animals were randomly divided into six experimental groups: SHAM-operated Wistar rats (W - SHAM), Wistar rats with induced postischemic AKI (W - AKI) and Wistar group with HBO preconditioning before AKI induction (W - AKI + HBO). On the other hand, SHR rats were also divided into same three groups: SHR - SHAM, SHR - AKI and SHR - AKI + HBO. We demonstrated that HBO preconditioning upregulated HO-1 and anti-apoptotic Bcl-2 protein expression, in both Wistar and SH rats. In addition, HBO preconditioning improved glomerular filtration rate, supporting by significant increase in creatinine, urea and phosphate clearances in both rat strains. Considering our results, we can also say that even in hypertensive conditions, we can expect protective effects of HBO preconditioning in experimental model of AKI.
... The antioxidant genes activated by HBOT include heme oxygenase-1 and metallothionein, which have been reported to protect cells from oxidative stress-induced damage (Bell and Vallee 2009;Xue et al. 2009;Verma et al. 2010;Hou et al. 2012). The ability of HBOT to activate the expression of antioxidant genes has also been reported for a number of other cell and tissue types (Shiraishi et al. 1983;Padgaonkar et al. 1997;Dennog et al. 1999;Rothfuss et al. 2001). However, little is known about the functional consequences of these gene expression changes and whether HBOT can indeed protect tissues from oxidative damage. ...
Article
Full-text available
Hyperbaric oxygen therapy (HBOT) is used for a number of applications, including the treatment of diabetic foot ulcers and CO poisoning. However, we and others have shown that HBOT can mobilize cellular antioxidant defenses, suggesting that it may also be useful under circumstances in which tissue protection from oxidative damage is desired. To test the protective properties of hyperbaric oxygen (HBO) on a tissue level, we evaluated the ability of a preconditioning treatment regimen to protect cutaneous tissue from UV-A-induced oxidative damage. Three groups of hairless SKH1-E mice were exposed to UV-A 3 days per week for 22 weeks, with two of these groups receiving an HBO pretreatment either two or four times per week. UV-A exposure increased apoptosis and proliferation of the skin tissue, indicating elevated levels of epithelial damage and repair. Pretreatment with HBO significantly reduced UV-A-induced apoptosis and proliferation. A morphometric analysis of microscopic tissue folds also showed a significant increase in skin creasing following UV-A exposure, which was prevented by HBO pretreatment. Likewise, skin elasticity was found to be greatest in the group treated with HBO four times per week. The effects of HBO were also apparent systemically as reductions in caspase-3 activity and expression were observed in the liver. Our findings support a protective function of HBO pretreatment from a direct oxidative challenge of UV-A to skin tissue. Similar protection of other tissues may likewise be achievable.
Article
Full-text available
In the present study, we investigated the effects of exhaustive exercise and recovery on inflammatory, pro-apoptotic, and anti-oxidative responses in human peripheral blood mononuclear cells (PBMCs). Sixteen volunteers participated in a guided physical activity program in which they were subjected to progressive exercise on the treadmill until they were exhausted followed by an 1-hour recovery period. Isolated human PBMCs were collected before exercise, immediately after exercise, and after 1-hour recovery. Exhaustive exercise induced expression of heme oxygenase-1 and glutamate cysteine ligase catalytic subunit and activation of NF-κB and NF-E2 related factor 2 (Nrf2). Apoptosis, as measured by activity and cleavage of caspase-3 and its substrate PARP also significantly increased. However, induction of redox signaling and the pro-apoptotic response fully returned to the baseline level during the 1-hour recovery period. On the other hand, COX-2 expression was continuously elevated after exercise cessation throughout the 1-hour recovery period. Taking all these findings into account, we conclude that exhaustive exercise transiently induces Nrf2-mediated antioxidant gene expression and eliminates damaged cells through apoptosis as part of an adaptive cytoprotective response against oxidative and inflammatory stress.
Article
Full-text available
Nonhealing diabetic wounds are common complications for diabetic patients. Because chronic hypoxia prominently delays wound healing, sustained oxygenation to alleviate hypoxia is hypothesized to promote diabetic wound healing. However, sustained oxygenation cannot be achieved by current clinical approaches, including hyperbaric oxygen therapy. Here, we present a sustained oxygenation system consisting of oxygen-release microspheres and a reactive oxygen species (ROS)–scavenging hydrogel. The hydrogel captures the naturally elevated ROS in diabetic wounds, which may be further elevated by the oxygen released from the administered microspheres. The sustained release of oxygen augmented the survival and migration of keratinocytes and dermal fibroblasts, promoted angiogenic growth factor expression and angiogenesis in diabetic wounds, and decreased the proinflammatory cytokine expression. These effects significantly increased the wound closure rate. Our findings demonstrate that sustained oxygenation alone, without using drugs, can heal diabetic wounds.
Article
Full-text available
Hyperbaric oxygen therapy (HBOT) is commonly used as treatment in several diseases, such as non-healing chronic wounds, late radiation injuries and carbon monoxide poisoning. Ongoing research into HBOT has shown that preconditioning for surgery is a potential new treatment application, which may reduce complication rates and hospital stay. In this review, the effect of HBOT on oxidative stress, inflammation and angiogenesis is investigated to better understand the potential mechanisms underlying preconditioning for surgery using HBOT. A systematic search was conducted to retrieve studies measuring markers of oxidative stress, inflammation, or angiogenesis in humans. Analysis of the included studies showed that HBOT-induced oxidative stress reduces the concentrations of pro-inflammatory acute phase proteins, interleukins and cytokines and increases growth factors and other pro-angiogenesis cytokines. Several articles only noted this surge after the first HBOT session or for a short duration after each session. The anti-inflammatory status following HBOT may be mediated by hyperoxia interfering with NF-κB and IκBα. Further research into the effect of HBOT on inflammation and angiogenesis is needed to determine the implications of these findings for clinical practice.
Article
The aim of the study was to evaluate the effects of dietary leucine level on growth performance, intestinal antioxidant status and immune response involved in Nrf2 and NF-κB signaling pathway in juvenile golden pompano (Trachinotus ovatus). A total of 450 juvenile golden pompano (9.15 ± 0.04 g) were fed three isonitrogenous diets with graded leucine levels [1.25% (control), 2.77% and 5.84%] for 8 weeks. The results showed that, compared with the control group, the WG was significantly improved in fish fed with 2.77% of dietary leucine (P < 0.05), and the 5.84% dietary leucine group had a tendency to increase. Compared to control group, 5.84% dietary leucine group significantly decreased the moisture and ash contents of whole body (P < 0.05), meanwhile, 2.77% dietary leucine group significantly decreased moisture content of whole body, but significantly improved the whole body crude lipid content (P < 0.05). Compared with the control group, the ALP level was significantly improved in fish fed with 2.77% of dietary leucine (P < 0.05). Inversely, the AST and ALT activities were significantly decreased in fish fed with 2.77% dietary leucine level (P < 0.05). Compared with the control group, GPx, T-AOC, SOD activities in group of 2.77% dietary arginine level were significantly increased (P < 0.05). However, MDA level showed a reverse trend, which was significantly decreased in fish fed with 2.77% dietary leucine level (P < 0.05). 2.77% dietary leucine levels significantly increased the relative expressions of Nrf2, HO-1, Cu/Zn-SOD, Mn-SOD and CAT (P < 0.05). In contrast, the relative expression of Keap1 showed a converse trend. Compared with the control group, the relative expressions of NF-κB, TNF-α and IL1-β were significantly lowered in fish fed with 2.77% of dietary leucine (P < 0.05). Additionally, 2.77% dietary leucine level significantly improved the relative expressions of TGF-β and IL-10 (P < 0.05). The 2.77% dietary leucine level significantly increased the muscular thickness compared with 5.84% dietary leucine level (P < 0.05). Furthermore, compared with the control group, the villus height and goblet cell counts were significantly improved in fish fed with 2.77% of dietary leucine (P < 0.05). These results indicated that the optimum dietary leucine plays an important role in promoting growth, enhancing antioxidant and immunity to maintain the intestinal health status of juvenile golden pompano.
Article
Full-text available
Postoperative cognitive dysfunction is a crucial public health issue that has been increasingly studied in efforts to reduce symptoms or prevent its occurrence. However, effective advances remain lacking. Hyperbaric oxygen preconditioning has proved to protect vital organs, such as the heart, liver, and brain. Recently, it has been introduced and widely studied in the prevention of postoperative cognitive dysfunction, with promising results. However, the neuroprotective mechanisms underlying this phenomenon remain controversial. This review summarizes and highlights the definition and application of hyperbaric oxygen preconditioning, the perniciousness and pathogenetic mechanism underlying postoperative cognitive dysfunction, and the effects that hyperbaric oxygen preconditioning has on postoperative cognitive dysfunction. Finally, we conclude that hyperbaric oxygen preconditioning is an effective and feasible method to prevent, alleviate, and improve postoperative cognitive dysfunction, and that its mechanism of action is very complex, involving the stimulation of endogenous antioxidant and anti-inflammation defense systems.
Chapter
Hyperbaric oxygen treatment (HBOT) has been reported to improve cerebral hypoperfusion, inflammation, and mitochondrial dysfunction, all of which have been observed in some individuals with autism spectrum disorders (ASD). In several studies, HBOT improved cerebral hypoperfusion, decreased a marker of inflammation, and did not worsen oxidative stress markers at pressures commonly used in children with ASD. Most studies of HBOT in children with ASD reported improvements in several behavioral domains, although many of these studies were not controlled. The two studies employing a control group appear to have conflicting results. However, this may have been due to different sample sizes and frequencies of HBOT sessions, with the larger study reporting improvements in several domains using a greater intensity of treatments. All studies reported that HBOT had minimal adverse events. Additional studies of HBOT in individuals with ASD are warranted to determine optimal treatment protocols.
Article
A major generalized and unspecific defence mechanism involves the expression of stress proteins, both at the cellular and organ level. Stress proteins are primarily transcriptionally regulated and inducible by a variety of inducers, such as oxidative stress or inflammation. The stress proteins most investigated so far are the heat shock protein 70 (HSP70) and the heme oxygenase-1 (HO-1, HSP32). Apart from their functional importance in sublethal stress of parts of the organism, stress proteins have an enormous protective capacity of considerable therapeutic potential, in particular because of their antioxidative, anti-inflammatory and vasodilative effects. Nearly all cells are basically capable of increasing their own resistance to recurring damaging influences. However, this acquired state of "stress tolerance" is transient and limited to a narrow period of time and can only be induced under physiological conditions. Stress conditioning, i.e. preinduction of the stress response, permits a protection of organs and organisms from further damage. This can be observed in hyperoxic lung injury, reperfusion injury subsequent to ischaemia, organ integrity after transplantation, or ischaemic preconditioning.
Article
Full-text available
The disease burden from diabetic kidney disease is large and growing. Effective therapies are lacking, despite an urgent need. Hyperbaric oxygen therapy (HBOT) activates Nrf2 and cellular antioxidant defenses; therefore, it may be generally useful for treating conditions that feature chronic oxidative tissue damage. Herein, we determined how periodic exposure to oxygen at elevated pressure affected type 2 diabetes mellitus-related changes in the kidneys of db/db mice. Two groups of db/db mice, designated 2.4 ATA and 1.5 ATA, were treated four times per week with 100 % oxygen at either 1.5 or 2.4 ATA (atmospheres absolute) followed by tests to assess kidney damage and function. The sham group of db/db mice and the Hets group of db/+ mice were handled but did not receive HBOT. Several markers of kidney damage were reduced significantly in the HBOT groups including urinary biomarkers neutrophil gelatinase-associated lipocalin (NGAL) and cystatin C (CyC) along with significantly lower levels of caspase-3 activity in kidney tissue extracts. Other stress biomarkers also showed trends to improvement in the HBOT groups, including urinary albumin levels. Expressions of the stress response genes NRF2, HMOX1, MT1, and HSPA1A were reduced in the HBOT groups at the end of the experiment, consistent with reduced kidney damage in treated mice. Urinary albumin/creatinine ratio (ACR), a measure of albuminuria, was significantly reduced in the db/db mice receiving HBOT. All of the db/db mouse groups had qualitatively similar changes in renal histopathology. Glycogenated nu-clei, not previously reported in db/db mice, were observed in these three experimental groups but not in the control group of nondiabetic mice. Overall, our findings are consistent with therapeutic HBOT alleviating stress and damage in the diabetic kidney through cytoprotective responses. These findings support an emerging paradigm in which tissue oxygenation and cellular defenses effectively limit damage from chronic oxida-tive stress more effectively than chemical antioxidants.
Article
Despite established exposure limits and safety standards, and the availability of carbon monoxide (CO) alarms, each year 50,000 people in the United States visit emergency departments for CO poisoning. Carbon monoxide poisoning can occur from brief exposures to high levels of CO, or from longer exposures to lower levels. Common symptoms include headaches, nausea and vomiting, dizziness, general malaise, and altered mental status. Some patients may have chest pain, shortness of breath and myocardial ischemia, and may require mechanical ventilation and treatment of shock. Individuals poisoned by CO often go on to develop neurological problems, including cognitive sequelae, anxiety and depression, persistent headaches, dizziness, sleep problems, motor weakness, vestibular and balance problems, gaze abnormalities, peripheral neuropathies, hearing loss, tinnitus and Parkinsonian-like syndrome. While breathing oxygen hastens the removal of carboxyhemoglobin (COHb), hyperbaric oxygen (HBO2) hastens COHb elimination and favorably modulates inflammatory processes instigated by CO poisoning, an effect not observed with breathing normobaric oxygen. Hyperbaric oxygen improves mitochondrial function, inhibits lipid peroxidation transiently, impairs leukocyte adhesion to injured microvasculature, and reduces brain inflammation caused by the CO-induced adduct formation of myelin basic protein. Based upon three supportive randomized clinical trials in humans and considerable evidence from animal studies, HBO2 should be considered for all cases of acute symptomatic CO poisoning. Hyperbaric oxygen is indicated for CO poisoning complicated by cyanide poisoning, often concomitantly with smoke inhalation.
Article
Full-text available
Testicular torsion is a urological emergency most commonly seen in adolescence, involving a decrease in blood flow in the testis resulting from torsion of the spermatic cord that can result in gonad injury or even loss if not treated in time. Testicular ischaemia-reperfusion injury represents the principle pathophysiology of testicular torsion, with ischaemia caused by twisting of the spermatic cord, and reperfusion on its subsequent release. Many cellular and molecular mechanisms are involved in ischaemia-reperfusion injury following testicular torsion. Studies have investigated the use of pharmacological agents as supportive therapy to surgical repair in order to prevent the adverse effects of testicular torsion. Numerous substances have been proposed as important in the prevention of post-ischaemia-reperfusion testicular injury. A range of chemicals and drugs has been successfully tested in animal models for the purpose of mitigating the dangerous effects of ischaemia-reperfusion in testis torsion.
Article
Significance: Oxidative stress is recognized as playing a role in stem cell mobilization from peripheral sites and also cell function. Recent advances: This review focuses on the impact of hyperoxia on vasculogenic stem cells and elements of wound healing. Critical issues: Components of the wound-healing process in which oxidative stress has a positive impact on the various cells involved in wound healing are highlighted. A slightly different view of wound-healing physiology is adopted by departing from the often used notion of sequential stages: hemostatic, inflammatory, proliferative, and remodeling and instead organizes the cascade of wound healing as overlapping events or waves pertaining to reactive oxygen species, lactate, and nitric oxide. This was done because hyperoxia has effects of a number of cell signaling events that converge to influence cell recruitment/chemotaxis and gene regulation/protein synthesis responses which mediate wound healing. Future directions: Our alternative perspective of the stages of wound healing eases recognition of the multiple sites where oxidative stress has an impact on wound healing. This aids the focus on mechanistic events and the interplay among various cell types and biochemical processes. It also highlights the areas where additional research is needed.
Article
Background Hyperbaric oxygen therapy (HBOT) provides 100% oxygen under pressure, which increases tissue oxygen levels, relieves hypoxia and alters inflammatory pathways. Although there is experience using HBOT in Crohn's disease and ulcerative colitis, the safety and overall efficacy of HBOT in inflammatory bowel disease (IBD) is unknown.AimTo quantify the safety and efficacy of HBOT for Crohn's disease (CD) and ulcerative colitis (UC). The rate of adverse events with HBOT for IBD was compared to the expected rate of adverse events with HBOT.MethodsMEDLINE, EMBASE, Cochrane Collaboration and Web of Knowledge were systematically searched using the PRISMA standards for systematic reviews. Seventeen studies involving 613 patients (286 CD, 327 UC) were included.ResultsThe overall response rate was 86% (85% CD, 88% UC). The overall response rate for perineal CD was 88% (18/40 complete healing, 17/40 partial healing). Of the 40 UC patients with endoscopic follow-up reported, the overall response rate to HBOT was 100%. During the 8924 treatments, there were a total of nine adverse events, six of which were serious. The rate of adverse events with HBOT in IBD is lower than that seen when utilising HBOT for other indications (P < 0.01). The risk of bias across studies was high.Conclusions Hyperbaric oxygen therapy is a relatively safe and potentially efficacious treatment option for IBD patients. To understand the true benefit of HBOT in IBD, well-controlled, blinded, randomised trials are needed for both Crohn's disease and ulcerative colitis.
Article
Adjunctive hyperbaric oxygen therapy is a safe and effective modality with which to increase tissue oxygenation and aid in healing of difficult wounds. The majority of the literature surrounding hyperbaric oxygen therapy supports its use in chronic wounds, but its use in acute wounds, flaps, and grafts is less well supported. The authors reviewed the Ovid, PubMed, and Cochrane Library databases, and selected studies, level III and above, using hyperbaric oxygen therapy in the treatment of complicated acute wounds, flaps, and grafts. A total of eight studies were found to meet criteria for evaluation of adjunctive hyperbaric oxygen therapy in the treatment of complicated acute wounds, flaps, and grafts. When combined with standard wound management principles, hyperbaric oxygen therapy can augment healing in complicated acute wounds. However, it is not indicated in normal wound management. Further investigation is required before it can be recommended as a mainstay in adjuvant wound therapy. Therapeutic, II.
Article
Full-text available
Oxygen treatment has been a cornerstone of acute medical care for numerous pathological states. Initially, this was supported by the assumed need to avoid hypoxaemia and tissue hypoxia. Most acute treatment algorithms, therefore, recommended the liberal use of a high fraction of inspired oxygen, often without first confirming the presence of a hypoxic insult. However, recent physiological research has underlined the vasoconstrictor effects of hyperoxia on normal vasculature and, consequently, the risk of significant blood flow reduction to the at-risk tissue. Positive effects may be claimed simply by relief of an assumed local tissue hypoxia, such as in acute cardiovascular disease, brain ischaemia due to, for example, stroke or shock or carbon monoxide intoxication. However, in most situations, a generalized hypoxia is not the problem and a risk of negative hyperoxaemia-induced local vasoconstriction effects may instead be the reality. In preclinical studies, many important positive anti-inflammatory effects of both normobaric and hyperbaric oxygen have been repeatedly shown, often as surrogate end-points such as increases in gluthatione levels, reduced lipid peroxidation and neutrophil activation thus modifying ischaemia-reperfusion injury and also causing anti-apoptotic effects. However, in parallel, toxic effects of oxygen are also well known, including induced mucosal inflammation, pneumonitis and retrolental fibroplasia. Examining the available 'strong' clinical evidence, such as usually claimed for randomized controlled trials, few positive studies stand up to scrutiny and a number of trials have shown no effect or even been terminated early due to worse outcomes in the oxygen treatment arm. Recently, this has led to less aggressive approaches, even to not providing any supplemental oxygen, in several acute care settings, such as resuscitation of asphyxiated newborns, during acute myocardial infarction or after stroke or cardiac arrest. The safety of more advanced attempts to deliver increased oxygen levels to hypoxic or ischaemic tissues, such as with hyperbaric oxygen therapy, is therefore also being questioned. Here, we provide an overview of the present knowledge of the physiological effects of oxygen in relation to its therapeutic potential for different medical conditions, as well as considering the potential for harm. We conclude that the medical use of oxygen needs to be further examined in search of solid evidence of benefit in many of the current clinical settings in which it is routinely used.
Article
Full-text available
Background/aims: This study investigated the effects of zinc on heme oxygenase-1 (HO-1) expression in human cancer cells. Methods/results: Zinc at sub-cytotoxic concentrations (50-100 μM) induces HO-1 expression in the MDA-MB-231 (human breast cancer) and A2780 (human ovarian cancer) cell lines in a concentration- and time-dependent manner. The induction of HO-1 by zinc was detected after 4-6 hours of treatment, reached maximal level at 8 hours, and declined thereafter. Using a human HO-1 gene promoter reporter construct, we identified two antioxidant response elements (AREs) that mediated the zinc-induced increase in HO-1 gene transcription, indicating that the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathway is involved in this event. This assumption was supported by the observations that knockdown of Nrf2 expression compromised the zinc-induced increase in HO-1 gene transcription, and that zinc increased Nrf2 protein expression and the Nrf2 binding to the AREs. Additionally, we found that the zinc-induced HO-1 gene transcription can be enhanced by clioquinol, a zinc ionophore, and reversed by pretreatment with TPEN, a known zinc chelator, indicating that an increase in intracellular zinc levels is responsible for this induction. Conclusion: These findings demonstrate that zinc at sub-cytotoxic concentrations induces HO-1 expression in human cancer cells. The biological significance of this induction merits further investigation.
Article
Oxidative stress induces the expression of the cytoprotective and anti-inflammatory protein haem oxygenase-1 (HO-1). In the present investigation, we show that anticipation of subsequent exercise elevates the expression of HO-1 mRNA in lymphocytes. A between-groups comparison of HO-1 mRNA expression in subjects about to complete a half marathon race vs. subjects who were asked to sit quietly in the laboratory showed an elevated expression of HO-1 mRNA prior to exercise (2.6-fold higher in subjects prior to the half marathon, P < 0.01). This observation led us to examine whether anticipation of subsequent exercise leads to differences in lymphocyte HO-1 mRNA expression within the same subjects. In a second experiment, the same individuals completed two trials, one exercise and one rest, approximately 2 weeks apart in a randomised cross-over design. Lymphocyte HO-1 mRNA expression was greater prior to exercise (1.4 ± 0.3-fold higher in the exercise trial, P < 0.05). These results suggest that knowledge of subsequent demanding exercise may lead to an anticipatory induction of HO-1 mRNA. We tentatively propose that this process has evolved to prepare lymphocytes for subsequent exercise-induced oxidative stress although the mechanism remains to be elucidated.
Article
Full-text available
Traditionally, hyperbaric oxygen treatment (HBOT) is indicated in several clinical disorders include decompression sickness, healing of problem wounds and arterial gas embolism. However, some investigators have used HBOT to treat individuals with autism spectrum disorders (ASD). A number of individuals with ASD possess certain physiological abnormalities that HBOT might ameliorate, including cerebral hypoperfusion, inflammation, mitochondrial dysfunction and oxidative stress. Studies of children with ASD have found positive changes in physiology and/or behavior from HBOT. For example, several studies have reported that HBOT improved cerebral perfusion, decreased markers of inflammation and did not worsen oxidative stress markers in children with ASD. Most studies of HBOT in children with ASD examined changes in behaviors and reported improvements in several behavioral domains although many of these studies were not controlled. Although the two trials employing a control group reported conflicting results, a recent systematic review noted several important distinctions between these trials. In the reviewed studies, HBOT had minimal adverse effects and was well tolerated. Studies which used a higher frequency of HBOT sessions (e.g., 10 sessions per week as opposed to 5 sessions per week) generally reported more significant improvements. Many of the studies had limitations which may have contributed to inconsistent findings across studies, including the use of many different standardized and non-standardized instruments, making it difficult to directly compare the results of studies or to know if there are specific areas of behavior in which HBOT is most effective. The variability in results between studies could also have been due to certain subgroups of children with ASD responding differently to HBOT. Most of the reviewed studies relied on changes in behavioral measurements, which may lag behind physiological changes. Additional studies enrolling children with ASD who have certain physiological abnormalities (such as inflammation, cerebral hypoperfusion, and mitochondrial dysfunction) and which measure changes in these physiological parameters would be helpful in further defining the effects of HBOT in ASD.
Article
Full-text available
Hyperbaric oxygen (HBO) therapy is used for the treatment of a variety of diseases, but also leads to oxidative stress as a result of increased formation of reactive oxygen species. The consequences may be damage to the lung, the central nervous system and the genome. The oxidative attack on DNA causes, among other damage, single and double strand breaks. Using the comet assay, a well-established genotoxicity test, it was possible to show that a single HBO exposure leads to increased levels of DNA strand breaks in a close dose-effect relationship. On the other hand, it was possible to demonstrate that these strand breaks are repaired rapidly and that, in repeated HBO exposures, DNA strand breaks occur only after the first treatment, not subsequent ones, indicating an induction of protective mechanisms. In healthy organisms, DNA repair and antioxidant mechanisms maintain a steady-state level of damage with minimal risk to the cell or the whole organism, but it cannot be excluded that HBO might lead to a significant mutational burden in situations where antioxidant defence is deficient or overwhelmed. The administration of antioxidants draws an ambivalent picture; Vitamin C, E or even N-acetylcysteine seems to be ineffective to prevent HBO-induced genotoxicity, whereas the orally effective vegetal superoxide dismutase (SOD, Glisodin®) is effective, and, thus, may play a role in the prevention of oxidative DNA damage.
Chapter
HBO has a DNA-damaging and possibly mutagenic potential. In vitro studies with mammalian cells suggest that HBO-induced oxidative DNA damage mainly leads to gross genetic alterations and chromosome aberrations. Under therapeutic exposure conditions, DNA damage is detected with the comet assay, but mutations and chromosome aberrations are not observed in peripheral blood cells. A very simple and efficient way to avoid HBO-induced DNA damage is to start with a shortened treatment before the standard protocol is applied the following days. Therefore, the hyperbaric community should think about an adaptation of the commonly used treatment protocols
Article
Both the phosphatidylinositol 3-kinase (PI3K)/Akt pathway and heme oxygenase-1 (HO-1) create a survival signal against oxidative stress-induced injuries. Although we have demonstrated that hydrogen peroxide (H2O2) preconditioning confers adaptive cytoprotection against oxidative stress-induced injury in PC12 cells, it remains unknown whether these defense systems are involved in the protective effect of H2O2 preconditioning. In the current study, PC12 cells were preconditioned with 100 µM H2O2 for 90 min, followed by 24 h recovery and subsequent exposure to 300 µM H2O2 for further 12 h. The findings showed that preconditioning with 100 µM H2O2 upregulated HO-1 expression. Zinc protoporphyrin IX (ZnPP), a selective inhibitor of HO-1, at a concentration of 15 µM, significantly attenuated H2O2 preconditioning-elicited cytotoxicity, apoptosis, oxidative stress and mitochondrial membrane potential (ΔΨm) loss in PC12 cells. In addition, H2O2 preconditioning enhanced phosphorylation of Akt. Treatment with 25 µM LY294002, a selective inhibitor of PI3K, for 20 min before H2O2 preconditioning blocked not only H2O2 preconditioning-induced HO-1 induction, but also the protective effect of H2O2 preconditioning against cytotoxicity. The present study provides novel evidence for the effect of preconditioning with H2O2 on the induction of HO-1, which contributes to the adaptive cytoprotection of H2O2 preconditioning against oxidative stress-induced cellular injury via a PI3K/Akt-dependent mechanism in PC12 cells.
Article
Full-text available
HSP72 is rapidly expressed in response to a variety of stressors in vitro and in vivo (including hypoxia). This project sought a hypoxic stimulus to elicit increases in HSP72 and HSP32 in attempts to confer protection to the sub-maximal aerobic exercise-induced disturbances to redox balance. Eight healthy recreationally active male subjects were exposed to five consecutive days of once-daily hypoxia (2,980 m, 75 min). Seven days prior to the hypoxic acclimation period, subjects performed 60 min of cycling on a cycle ergometer (exercise bout 1-EXB1), and this exercise bout was repeated 1 day post-cessation of the hypoxic period (exercise bout 2-EXB2). Blood samples were taken immediately pre- and post-exercise and 1, 4 and 8 h post-exercise for HSP72 and immediately pre, post and 1 h post-exercise for HSP32, TBARS and glutathione [reduced (GSH), oxidised (GSSG) and total (TGSH)], with additional blood samples obtained immediately pre-day 1 and post-day 5 of the hypoxic acclimation period for the same indices. Monocyte-expressed HSP32 and HSP72 were analysed by flow cytometry, with measures of oxidative stress accessed by commercially available kits. There were significant increases in HSP72 (P < 0.001), HSP32 (P = 0.03), GSSG (t = 9.5, P < 0.001) and TBARS (t = 5.6, P = 0.001) in response to the 5-day hypoxic intervention, whereas no significant changes were observed for GSH (P = 0.22) and TGSH (P = 0.25). Exercise-induced significant increases in HSP72 (P < 0.001) and HSP32 (P = 0.003) post-exercise in EXB1; this response was absent for HSP72 (P ≥ 0.79) and HSP32 (P ≥ 0.99) post-EXB2. The hypoxia-mediated increased bio-available HSP32 and HSP72 and favourable alterations in glutathione redox, prior to exercise commencing in EXB2 compared to EXB1, may acquiesce the disturbances to redox balance encountered during the second physiologically identical exercise bout.
Article
Full-text available
Traditionally, hyperbaric oxygen treatment (HBOT) has been used to treat a limited repertoire of disease, including decompression sickness and healing of problem wounds. However, some investigators have used HBOT to treat inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis. Comprehensive searches were conducted in 8 scientific databases through 2011 to identify publications using HBOT in IBD. Human studies and animal models were collated separately. Thirteen studies of HBOT in Crohn's disease and 6 studies in ulcerative colitis were identified. In all studies, participants had severe disease refractory to standard medical treatments, including corticosteroids, immunomodulators and anti-inflammatory medications. In patients with Crohn's disease, 31/40 (78%) had clinical improvements with HBOT, while all 39 patients with ulcerative colitis improved. One study in Crohn's disease reported a significant decrease in proinflammatory cytokines (IL-1, IL-6 and TNF-alpha) and one study in ulcerative colitis reported a decrease in IL-6 with HBOT. Adverse events were minimal. Twelve publications reported using HBOT in animal models of experimentally-induced IBD, including several studies reporting decreased markers of inflammation or immune dysregulation, including TNF-alpha (3 studies), IL-1beta (2 studies), neopterin (1 study) and myeloperoxidase activity (5 studies). HBOT also decreased oxidative stress markers including malondialdehyde (3 studies) and plasma carbonyl content (2 studies), except for one study that reported increased plasma carbonyl content. Several studies reported HBOT lowered nitric oxide (3 studies) and nitric oxide synthase (3 studies) and one study reported a decrease in prostaglandin E2 levels. Four animal studies reported decreased edema or colonic tissue weight with HBOT, and 8 studies reported microscopic improvements on histopathological examination. Although most publications reported improvements with HBOT, some studies suffered from limitations, including possible publication and referral biases, the lack of a control group, the retrospective nature and a small number of participants. HBOT lowered markers of inflammation and oxidative stress and ameliorated IBD in both human and animal studies. Most treated patients were refractory to standard medical treatments. Additional studies are warranted to investigate the effects of HBOT on biomarkers of oxidative stress and inflammation as well as clinical outcomes in individuals with IBD.
Article
Hyperbaric oxygen (HBO) treatment is based on the principle of having the patient breath 100% oxygen in an environment above atmospheric pressure. Ozone (O(3)) is a colourless gas with a specific odour and consists of three oxygen atoms. The classical scientific understanding is that the world has become a place suitable for life for aerobic organisms with the increasing oxygen in the atmosphere billions of years ago. The formation of ozone after oxygen has then protected aerobic creatures from harmful rays. We now use these two gases for treatment purposes. It is noteworthy that the oxygen and ozone molecules that are formed by the same atom in different numbers are used for similar medical indications. We will try to emphasize the similarities and differences of HBO and medical ozone applications in this article.
Article
Renal ischemia/reperfusion (I/R) injury occurs in both native and transplanted kidneys. Hyperbaric oxygen (HBO) has been shown to prevent I/R injury in different tissues. The aim of this study was to evaluate the effect of HBO on renal I/R injury in rats. Male Sprague-Dawley rats were randomly assigned to three groups. The sham group (n = 8) received right nephrectomy. The I/R (n = 8) and HBO + I/R groups (n = 8) received 45 min left renal ischemia followed by 24 h of reperfusion after right nephrectomy. The HBO + I/R group (n = 8) received 100% oxygen at 2.5 atmosphere absolute (ATA), for 1 h at every 12 h interval for 2 d. Reperfusion was performed 24 h later after the last HBO exposure. In HBO + I/R group, blood urea nitrogen (BUN) and creatinine levels decreased significantly compared with the sham and I/R groups (P < 0.01). Activities of superoxide dismutase (SOD) were increased in renal tissue in the HBO + I/R groups. The content of malondialdehyde (MDA) were decreased in the HBO + I/R groups. Kidney samples from HBO + I/R group rats revealed markedly reduced histological damage under histopathological examination. The animals treated with HBO showed significantly elevated heme oxygenase-1 (HO-1) protein and mRNA levels expression compared with I/R group (P < 0.05). Hyperbaric oxygen preconditioning (HBO-PC) can protect renal I/R injury against oxidative stress, and the up-regulation of HO-1 expression plays an essential role in HBO induced preconditioning effect.
Article
In vitro studies on hyperbaric oxygen (HBO) therapy suggest that HBO may cause DNA damage, but this has not been evaluated using endothelial cells. Human umbilical cord endothelial cells (HUVECs) were exposed either to H(2)O(2) or to HBO for 90 min, with or without subsequent H(2)O(2) exposure. Measurements included the comet assay for DNA damage, and reduced and oxidised glutathione levels. HUVECs showed sensitivity to H(2)O(2) (EC(50) of 0.2mM for DNA migration). A single 90 min HBO treatment at 2.2 ATA caused a statistically significant (ANOVA, P<0.05) increase of DNA migration in HUVECs to 6.8 ± 0.3% (mean ± SEM, n=8), which returned to normal levels (4.9 ± 0.1%, n=6) after 24h. Further exposure to 0.2mM H(2)O(2) after HBO treatment significantly increased the DNA migration in HBO-treated cells immediately post-treatment; but 24h later the cells showed 22% less DNA damage and higher glutathione than controls. A single HBO exposure causes limited DNA damage to HUVECs, which repairs quickly. HBO treatment protects against H(2)O(2)-induced DNA damage and involves cellular glutathione. Endothelial cells are unlikely to be compromised during HBO therapy.
Article
This article outlines therapeutic mechanisms of hyperbaric oxygen therapy and reviews data on its efficacy for clinical problems seen by plastic and reconstructive surgeons. The information in this review was obtained from the peer-reviewed medical literature. Principal mechanisms of hyperbaric oxygen are based on intracellular generation of reactive species of oxygen and nitrogen. Reactive species are recognized to play a central role in cell signal transduction cascades, and the discussion will focus on these pathways. Systematic reviews and randomized clinical trials support clinical use of hyperbaric oxygen for refractory diabetic wound-healing and radiation injuries; treatment of compromised flaps and grafts and ischemia-reperfusion disorders is supported by animal studies and a small number of clinical trials, but further studies are warranted. Clinical and mechanistic data support use of hyperbaric oxygen for a variety of disorders. Further work is needed to clarify clinical utility for some disorders and to hone patient selection criteria to improve cost efficacy.
Article
Hyperbaric oxygen (HBO) is thought to confer protection to cells via a cellular response to free radicals. This process may involve increased expression of heat shock proteins, in particular the highly inducible heat shock protein 72 (Hsp72). Healthy male volunteers (n = 16) were subjected to HBO for 1 h at 2.8 ATA. Inducible Hsp72 expression was measured by flow cytometry pre-, post- and 4 h-post HBO. Peripheral blood mononuclear cells (PBMC) were isolated from whole blood via density centrifugation pre-, post- and 4 h post-HBO. PBMC were then subjected to an in vitro heat shock at 40°C or hypoxia at 37°C (5% O(2)) with a control at 37°C. Cells were then analysed for Hsp72 expression by flow cytometry. Monocytes showed no significant changes in Hsp72 expression following HBO. No detectable Hsp72 was seen in lymphocytes or neutrophils. Following in vitro hypoxic exposure, a significant increase in Hsp72 expression was observed in monocytes isolated immediately post- (p = 0.006) and 4 h post-HBO (p = 0.010) in comparison to control values. HBO does not induce Hsp72 expression in PBMC. The reported benefits of HBO in terms of pre-conditioning are not due to inducement of Hsp72 expression in circulating blood cells, but may involve an enhancement of the stress response.
Article
The objective of this study was to investigate early effects of peritoneal inflammation on the mitochondrial function in the vital organs, liver and kidney, and their relation to inflammatory and oxidative stress mediators. The study was performed on 14 domestic pigs. Peritoneal inflammation was induced in anesthetized pigs after a midline laparotomy by autologous feces. Fluid resuscitation maintained a MAP above 60 mmHg. Animals were sacrificed 12 h later, and tissue samples were obtained to determine mitochondrial function, mRNA levels of relevant genes [inducible NO synthase (iNOS), inducible HO (HO-1), tumor necrosis factor-alpha (TNF-alpha)], generation of reactive oxygen species (ROS), and HO-1 activity. We found impaired mitochondrial function in both liver and kidney, based on decreased state 3 respiration in the liver and increased states 2 and 4 respiration in the kidney at 12 h. This was accompanied by increased TNF-alpha protein in the blood and up-regulation of TNF-alpha mRNA in the liver. Free iron was elevated in the liver but not in the kidney. In the kidney, mitochondrial ROS production was increased. Nitric oxide levels in blood remained unchanged, corresponding to unchanged levels of iNOS mRNA expression in liver and kidney. Similarly, HO-1 mRNA and heme oxygenase (HO)-activity were unchanged. The inflammatory response in the absence of characteristic septic symptoms was not associated with morphological organ damage at this early time point. Peritoneal inflammation in pigs caused mitochondrial dysfunction in liver and kidney, preceding signs of organ damage. We did not find proof that mitochondrial dysfunction was due to increased levels of either nitric oxide (NO) or products of HO, but it was accompanied by increased levels of oxidative stress markers.
Article
Full-text available
Recent reports that hyperbaric oxygenation (HBO2) induced apoptosis in T-cell lines raised concern about a possible immunosuppressive effect of HBO2. Nucleosomes, DNA fragments wrapped around a histone core, have been observed in the circulation in diseases with increased cell death such as sepsis. Our aim was to investigate, whether HBO2 increases circulating nucleosomes as a marker of cell death and induces apoptosis of peripheral blood mononuclear cells in vivo. After informed consent 29 healthy volunteers were exposed to a 30 minute dive at 2.8 atmospheres absolute in a pressure chamber under resting conditions, while breathing 100% oxygen. Samples were obtained before and 24 hours after exposure. Circulating nucleosomes were measured in serum. Caspase-3 activation, Bcl-2 expression and mRNA of Bcl-2, Bcl-xl and Bax were analyzed in mononuclear cell extracts. Nucleosomes were elevated markedly 24h after exposure (p<0.01), while caspase-3 was not activated significantly. mRNA levels of Bcl-2, Bcl-xl and Bax were not altered. In conclusion, while evidence of elevated levels of circulating nucleosomes was found, mononuclear cell apoptosis was not affected by a single exposure to hyperbaric oxygen.
Article
Full-text available
A single exposure to hyperbaric oxygen (HBO), i.e., pure oxygen breathing at supra-atmospheric pressures, causes oxidative DNA damage in humans in vivo as well as in isolated lymphocytes of human volunteers. These DNA lesions, however, are rapidly repaired, and an adaptive protection is triggered against further oxidative stress caused by HBO exposure. Therefore, we tested the hypothesis that long-term repetitive exposure to HBO would modify the degree of DNA damage. Combat swimmers and underwater demolition team divers were investigated because their diving practice comprises repetitive long-term exposure to HBO over years. Nondiving volunteers with and without endurance training served as controls. In addition to the measurement of DNA damage in peripheral blood (comet assay), blood antioxidant enzyme activities, and the ratio of oxidized and reduced glutathione content, we assessed the DNA damage and superoxide anion radical (O(2)(*-)) production induced by a single ex vivo HBO exposure of isolated lymphocytes. All parameters of oxidative stress and antioxidative capacity in vivo were comparable in the four different groups. Exposure to HBO increased both the level of DNA damage and O(2)(*-) production in lymphocytes, and this response was significantly more pronounced in the cells obtained from the combat swimmers than in all the other groups. However, in all groups, DNA damage was completely removed within 1 h. We conclude that, at least in healthy volunteers with endurance training, long-term repetitive exposure to HBO does not modify the basal blood antioxidant capacity or the basal level of DNA strand breaks. The increased ex vivo HBO-related DNA damage in isolated lymphocytes from these subjects, however, may reflect enhanced susceptibility to oxidative DNA damage.
Article
To gain a global view of the genomic response of neurons to normobaric and hyperbaric hyperoxic stress, we performed a microarray analysis of gene expression after exposure to varying levels of partial oxygen pressures. Rat neurons were exposed to normobaric hyperoxia, hyperbaric (2, 4, and 6 atmosphere absolute) air or hyperbaric O(2). We identified 183 genes significantly altered (increased or decreased >or=1.5-fold) in response to pressure and/or oxidative stress. Among them, 17 genes changed in response to all exposure conditions. More genes were altered in response to hyperbaric air than hyperbaric O(2). The altered genes included factors associated with stress responses, transport/neurotransmission, signal transduction, and transcription factors. The results may serve as guidance for selection of biomarkers of hyperoxia and hyperbaric O(2) response and provide a starting point for further studies to investigate the global molecular mechanisms underlying hyperbaric oxidative stress.
Article
The goal of this review is to outline advances addressing the role that reactive species of oxygen and nitrogen play in therapeutic mechanisms of hyperbaric oxygen. The review will be organized around major categories of problems or processes where controlled clinical trials have demonstrated clinical efficacy for hyperbaric oxygen therapy. Reactive species are now recognized to play a major role in cell signal transduction cascades, and the discussion will focus on how hyperbaric oxygen acts through these pathways to mediate wound healing and ameliorate postischemic and inflammatory injuries.
Article
Hyperbaric oxygen (HBO) treatment of cell cultures is a well suited model for studying genetic and cellular consequences of oxidative stress. We have previously shown that exposure of isolated human lymphocytes to HBO induces DNA damage and leads to the development of an adaptive response which protects lymphocytes from oxidative DNA damage induced by a repeated HBO exposure or by treatment with H(2)O(2). Our earlier studies also provided evidence for a functional involvement of the inducible enzyme heme oxygenase-1 (HO-1) in this adaptive protection. In contrast, V79 Chinese hamster cells did neither show a comparable adaptive protection nor an induction of HO-1 after HBO exposure. We now investigated possible mechanism(s) by which HO-1 contributes to an enhanced resistance of lymphocytes against oxidative stress. HO-1 catalyzes the rate-limiting step in heme degradation to form carbon monoxide (CO), biliverdin and free iron. We can now show that supplementation with exogenous CO does not protect V79 cells from HBO-induced oxidative DNA damage suggesting that increased generation of CO cannot account for the observed adaptive protection. On the other hand, HBO-exposed lymphocytes showed a small but reproducible increase in cellular ferritin levels, which might indicate that the underlying protective mechanism is based on an induction of ferritin, which may act antioxidatively by preventing the generation of the DNA-damaging hydroxyl radical via Fenton reaction. Our results further show that isolated lymphocytes also induce HO-1 and develop an adaptive protection when the first HBO exposure does not induce DNA damage, indicating that DNA damage is not the trigger for the development of the adaptive protection.
Article
Hyperbaric oxygen (HBO) treatment is applied as a therapy for a wide variety of diseases with symptoms caused by lack of oxygen in the target tissues. However, it is known that exposure to high concentrations of oxygen may lead to oxidative stress and cause cell and tissue damage. Oxygen toxicity and possible cancer-promoting effects of HBO therapy have been a matter of serious concern. Although a cancer-inducing effect of HBO was not found to date, recent studies clearly indicated an induction of oxidative DNA damage in blood cells of healthy subjects after HBO under therapeutic conditions. The biological significance of this finding has been investigated in a series of in vitro and in vivo tests. This review summarizes these studies and critically discusses potential adverse genetic effects of HBO therapy. Furthermore, since an induction of anti-oxidative defense mechanisms has been determined after HBO exposure, a modified treatment regimen of HBO therapy is proposed which avoids genotoxic effects.
Article
Heme oxygenase (HO) catalyzes the rate-limiting step in the oxidative degradation of heme to biliverdin. The isoform HO-1 is inducible by a variety of agents causing oxidative stress and has been suggested to play an important role in cellular protection against oxidant-mediated cell damage. Using treatment of cell cultures with hyperbaric oxygen (HBO) as a model for oxidative stress, we have shown an induction of HO-1 in isolated human lymphocytes after a single HBO exposure and protection of these cells against DNA damage by subsequent oxidative stress. In contrast, V79 Chinese hamster cells showed neither a comparable adaptive protection nor an induction of HO-1 after HBO exposure, which makes this cell line an attractive model system for a further characterization of HO-1-mediated protection. In the present study, we investigated whether overexpression of HO-1 renders V79 cells more resistant to DNA damage induced by HBO. Transient transfection of V79 cells with a full-length human HO-1 cDNA resulted in a 2-3-fold increase in HO-1 protein levels. Comet assay experiments with and without FPG posttreatment for the determination of oxidative DNA base damage showed that HO-1 overexpressing V79 cells were significantly protected against oxidative DNA damage induced by a single HBO exposure. Furthermore, HO-1-transfected cells exhibited a clearly reduced induction of micronuclei after HBO treatment. Since the observed protective effects were abolished by cotreatment with the HO-1 inhibitor tin-mesoporphyrin, our study suggests that a low-level overexpression of HO-1 provides protection against oxidative DNA damage induced by HBO.
Article
Full-text available
Accumulation of heme oxygenase mRNA is strongly stimulated by treatment of cultured human skin fibroblasts with ultraviolet radiation, hydrogen peroxide, or the sulfhydryl reagent sodium arsenite (S. M. Keyse and R. M. Tyrrell. Proc. Natl. Acad. Sci. USA, 86: 99-103, 1989). Since this will result in a transient reduction in the prooxidant state of cells, the phenomenon may represent an important inducible antioxidant defense mechanism. To examine the generality of the response, we have measured the accumulation of the specific mRNA in a variety of human and mammalian cell types after inducing treatments. Induction by sodium arsenite is observed in all additional human cell types tested. This includes primary epidermal keratinocytes and lung and colon fibroblasts as well as established cell lines such as HeLa, TK6 lymphoblastoid, and transformed fetal keratinocytes. Strong induction of heme oxygenase mRNA is also observed following sodium arsenite treatment of cell lines of rat, hamster, mouse, monkey, and marsupial origin. The agents which lead to induction in cultured human skin fibroblasts fall into two categories: (a) those which are oxidants or can generate active intermediates (ultraviolet A radiation, hydrogen peroxide, menadione, and the tumor promoter, 12-O-tetradecanoylphorbol-13-acetate); (b) agents which are known to interact with or modify cellular glutathione levels (buthionine sulfoximine, sodium arsenite, iodoacetamide, diamide, and cadmium chloride). These observations strongly support the hypothesis that induction of the enzyme is a general response to oxidant stress in mammalian cells and are consistent with the possibility that the cellular redox state plays a key role.
Article
Full-text available
Treatment of cultured human skin fibroblasts with near-UV radiation, hydrogen peroxide, and sodium arsenite induces accumulation of heme oxygenase mRNA and protein. In this study, these treatments led to a dramatic increase in the rate of RNA transcription from the heme oxygenase gene but had no effect on mRNA stability. Transcriptional activation, therefore, appears to be the major mechanism of stimulation of expression of this gene by either oxidative stress or sulfydryl reagents.
Article
Full-text available
Oxidative stress of human skin fibroblasts by treatment with ultraviolet A (UVA) radiation has been shown to lead to an increase in levels of the heme catabolizing enzyme heme oxygenase 1 [heme, hydrogen-donor:oxygen oxidoreductase (alpha-methene-oxidizing, hydroxylating), EC 1.14.99.3] and the iron storage protein ferritin. Here we show that human skin fibroblasts, preirradiated with UVA, sustain less membrane damage during a subsequent exposure to UVA radiation than cells that had not been preirradiated. Pretreating cells with heme oxygenase 1 antisense oligonucleotide inhibited the irradiation-dependent induction of both the heme oxygenase I enzyme and ferritin and abolished the protective effect of preirradiation. Inhibition of the UVA preirradiation-dependent increase in ferritin, but not heme oxygenase, with desferrioxamine also abolished the protection. This identifies heme oxygenase 1 as a crucial enzymatic intermediate in an oxidant stress-inducible antioxidant defense mechanism, involving ferritin, in human skin fibroblasts.
Article
Full-text available
Heme oxygenase-1 mRNA levels increase following exposure of many mammalian cell lines to oxidative stress such as ultraviolet A (UVA) irradiation. Here we demonstrate a 4-fold increase in microsomal heme oxygenase activity and a 40% decrease in microsomal heme content 14 h after treatment of human skin fibroblasts (FEK4) with 250 kJ m-2 of UVA radiation. Paralleling this was a 2-fold increase in ferritin levels that was sustained for at least 46 h after UVA irradiation. Treatment of fibroblasts with the iron chelating agent desferrioxamine, after the UVA-dependent induction of heme oxygenase, prevented the increase in ferritin levels. Treatment of fibroblasts with Sn-protoporphyrin IX (an inhibitor of heme oxygenase) also prevented the effect of UVA radiation on ferritin levels. Thus we conclude that the effect of UVA radiation on ferritin levels is via the heme oxygenase-dependent release of iron from endogenous heme sources. We propose that the increase in ferritin that follows UVA irradiation would decrease intracellular free iron such that iron-catalyzed free radical reactions would be restricted during periods of subsequent oxidative stress.
Article
Full-text available
A set of eight proteins is induced in peripheral blood lymphocytes from normal donors by exposure to hydrogen peroxide or to xanthine oxidase plus hypoxanthine. Four of them (hsp90, hsp72 and proteins 65 and 50 kDa) are also expressed after heat shock, together with proteins 110, 100 and 38 kDa. Among proteins induced after oxidative stress is a 32 kDa protein-probably corresponding to heme oxygenase-1 (HO-1)- and a 27 kDa protein, both known to be induced by reactive oxygen species. Although ionizing radiation is known to generate a number of pro-oxidant intermediates, using our one-dimensional electrophoresis system we can detect no differences in the proteins synthesized after exposure to gamma-ray doses between 5 and 20 Gy as compared with control cells. Pre-exposure to a mild hyperthermia or to moderate oxidative stress significantly increases survival of lymphocytes challenged with high doses of reactive oxygen species, in conditions compatible with a protective rôle exerted by stress proteins. The increase in survival is accompanied by the maintenance of the proliferative capacity of the cells. The physiological rôle played by stress proteins in prevention and repair of damage and the relationships between stress protein induction, oxidative state, proliferation and mode of cell death are discussed.
Article
Full-text available
Heme oxygenase (HO) catalyzes the rate-limiting step in the degradation of heme to biliverdin, which is reduced by biliverdin reductase to bilirubin. Heme oxygenase-1 (HO-1) is inducible not only by its heme substrate, but also by a variety of agents causing oxidative stress. Although much is known about the regulation of HO-1 expression, the functional significance of HO-1 induction after oxidant insult is still poorly understood. We hypothesize and provide evidence that HO-1 induction serves to protect cells against oxidant stress. Human pulmonary epithelial cells (A549 cells) stably transfected with the rat HO-1 cDNA exhibit marked increases of HO-1 mRNA levels which were correlated with increased HO enzyme activity. Cells that overexpress HO-1 (A549-A4) exhibited a marked decrease in cell growth compared with wild-type A549 (A549-WT) cells or A549 cells transfected with control DNA (A549-neo). This slowing of cell growth was associated with an increased number of cells in G0/G1 phase during the exponential growth phase and decreased entry into the S phase, as determined by flow cytometric analysis of propidium iodide-stained cells and pulse experiments with bromodeoxyuridine. Furthermore, the A549-A4 cells accumulated at the G2/M phase and failed to progress through the cell cycle when stimulated with serum, whereas the A549-neo control cells exhibited normal cell cycle progression. Interestingly, the A549-A4 cells also exhibited marked resistance to hyperoxic oxidant insult. Tin protoporphyrin, a selective inhibitor of HO, reversed the growth arrest and ablated the increased survival against hyperoxia observed in the A549-A4 cells overexpressing HO-1. Taken together, our data suggest that overexpression of HO-1 results in cell growth arrest, which may facilitate cellular protection against non-heme-mediated oxidant insult such as hyperoxia.
Article
Full-text available
Hyperbaric oxygen HBO therapy is successfully used for the treatment of a variety of conditions. However, exposure to high concentrations of oxygen is known to induce damage to cells, possibly due to an increased oxygen radical production. As reactive oxygen species also cause DNA damage, we investigated the DNA-damaging effect of HBO with the alkaline version of the single cell gel test comet assay. Oxidative DNA base modifications were determined by converting oxidized DNA bases to strand breaks using bacterial formamidopyrimidine-DNA glycosylase FPG, a DNA repair enzyme, which specifically nicks DNA at sites of 8-oxo-guanines and formamidopyrimidines. HBO treatment under therapeutic conditions clearly and reproducibly induced DNA damage in leukocytes of all test subjects investigated. Increased DNA damage was found immediately at the end of the treatment, while 24 h later, no effect was found. Using FPG protein we detected significant oxidative base damage after HBO treatment DNA damage was detected only after the first treatment and not after further treatments under the same conditions, indicating an increase in antioxidant defences. DNA damage did not occur when the HBO treatment was started with a reduced treatment time which was then increased stepwise.
Article
Full-text available
The role of heme oxygenase (HO)-1 was evaluated in the oxygen-resistant hamster fibroblast cell line, O2R95, which moderately overexpress HO when compared with the parental cell line, HA-1. To suppress HO-1 expression, O2R95 were transfected with HO-1 antisense oligonucleotide or treated with tin-mesoporphyrin (SnMP). To increase HO-1 expression, cells were transfected with HO-1 cDNA in a pRC/cytomegalovirus (CMV) vector. All cells were challenged with a 48-h exposure to 95% O2 (hyperoxia). When HO activity was suppressed, O2R95 cells had significantly decreased cell viability, increased susceptibility to lipid peroxidation, and increased protein oxidation in hyperoxia. In contrast, further overexpression of HO-1 did not improve resistance to oxygen toxicity. Antisense-transfected cells and SnMP-treated cells with lowered HO activity showed increased levels of cellular heme compared with controls. In the HO-1 cDNA-transfected O2R95 cells, cellular heme was lowered compared with controls; however, cellular redox active iron levels were increased. We conclude that HO mediates cytoprotection to oxygen toxicity within a narrow range of expression. We speculate that this protective effect may be mediated in part through increased metabolism of the pro-oxidant heme but that higher levels of HO activity obviate protection by increased redox active iron release.
Article
Full-text available
Hyperbaric oxygen (HBO) treatment (i.e. exposure to 100% oxygen at a pressure of 2.5 ATA for a total of three 20 min periods) of human subjects caused clear and reproducible DNA effects in the comet assay with leukocytes. Interestingly, DNA damage was detected only after the first treatment and not after further treatments under the same conditions, indicating an increase in antioxidant defences. We now demonstrate that blood taken 24 h after HBO treatment is well protected against the in vitro induction of DNA damage by hydrogen peroxide (H2O2). H2O2 treatment caused a significant induction of DNA effects in the comet assay and chromosome breakage in the micronucleus test in the blood of volunteers before HBO. The same treatment did not cause genotoxic effects 24 h after HBO. This protective effect lasted for at least 1 week. Experiments with isolated lymphocytes gave similar results, indicating that the adaptive response is a cellular effect. The cells were not comparably protected against the genotoxic effects of gamma-irradiation, suggesting increased scavenging of reactive oxygen species distant from nuclear DNA.
Article
Full-text available
Heme oxygenase (HO) is responsible for the physiological breakdown of heme into equimolar amounts of biliverdin, carbon monoxide, and iron. Three isoforms (HO-1, HO-2, and HO-3) have been identified. HO-1 is ubiquitous and its mRNA and activity can be increased several-fold by heme, other metalloporphyrins, transition metals, and stimuli that induce cellular stress. HO-1 is recognized as a major heat shock/stress response protein. Recent work from our laboratory has demonstrated several potential consensus regulatory elements in the 5'-untranslated region (UTR) of HO-1, including activator protein 1 (AP-1), metal responsive element (MRE), oncogene c-myc/max heterodimer binding site (Myc/Max), antioxidant response element (ARE), and GC box binding (Sp1) sites. Using deletion-reporter gene constructs, we have mapped sites that mediate the arsenite-dependent induction of HO-1, and we have shown that components of the extracellular signal-regulated kinase (ERK) and p38 (a homologue of the yeast HOG1 kinase), but not c-jun N-terminal kinase (JNK), mitogen-activated protein (MAP) kinase pathways are involved in arsenite-dependent upregulation. In contrast, HO-2 is present chiefly in the brain and testes and is virtually uninducible. HO-3 has very low activity; its physiological function probably involves heme binding. Products of the HO reaction have important effects: carbon monoxide is a potent vasodilator, which is thought to play a key role in the modulation of vascular tone, especially in the liver under physiological conditions, and in many organs under "stressful" conditions associated with HO-1 induction. Biliverdin and its product bilirubin, formed in most mammals, are potent antioxidants. In contrast, "free" iron increases oxidative stress and regulates the expression of many mRNAs (e.g., DCT-1, ferritin, and transferrin receptor) by affecting the conformation of iron regulatory protein (IRP)-1 and its binding to iron regulatory elements (IREs) in the 5'- or 3'-UTRs of the mRNAs.
Article
Full-text available
Haem oxygenase-1 (HO1) is a heat-shock protein that is induced by stressful stimuli. Here we demonstrate a cytoprotective role for HO1: cell death produced by serum deprivation, staurosporine or etoposide is markedly accentuated in cells from mice with a targeted deletion of the HO1 gene, and greatly reduced in cells that overexpress HO1. Iron efflux from cells is augmented by HO1 transfection and reduced in HO1-deficient fibroblasts. Iron accumulation in HO1-deficient cells explains their death: iron chelators protect HO1-deficient fibroblasts from cell death. Thus, cytoprotection by HO1 is attributable to its augmentation of iron efflux, reflecting a role for HO1 in modulating intracellular iron levels and regulating cell viability.
Article
Full-text available
The stress-inducible protein heme oxygenase-1 provides protection against oxidative stress. The anti-inflammatory properties of heme oxygenase-1 may serve as a basis for this cytoprotection. We demonstrate here that carbon monoxide, a by-product of heme catabolism by heme oxygenase, mediates potent anti-inflammatory effects. Both in vivo and in vitro, carbon monoxide at low concentrations differentially and selectively inhibited the expression of lipopolysaccharide-induced pro-inflammatory cytokines tumor necrosis factor-alpha, interleukin-1beta, and macrophage inflammatory protein-1beta and increased the lipopolysaccharide-induced expression of the anti-inflammatory cytokine interleukin-10. Carbon monoxide mediated these anti-inflammatory effects not through a guanylyl cyclase-cGMP or nitric oxide pathway, but instead through a pathway involving the mitogen-activated protein kinases. These data indicate the possibility that carbon monoxide may have an important protective function in inflammatory disease states and thus has potential therapeutic uses.
Article
Full-text available
Hyperbaric oxygen (HBO) treatment of human subjects (i.e. exposure to 100% oxygen at a pressure of 2.5 ATA for a total period of 3 x 20 min) caused clear and reproducible DNA damage in lymphocytes, as detected with the comet assay (single cell gel electrophoresis). Induction of DNA damage was found only after the first HBO exposure and not after further treatments of the same individuals. Furthermore, blood taken 24 h after HBO treatment was significantly protected against the induction of DNA damage by hydrogen peroxide (H(2)O(2)) in vitro, indicating that adaptation occurred due to induction of antioxidant defenses. The cells were not significantly protected against the genotoxic effects of gamma-irradiation, suggesting increased scavenging of reactive oxygen species distant from nuclear DNA or an inducible change in the levels of free transition metals. We now demonstrate increased levels of heme oxygenase-1 (HO-1) in lymphocytes 24 h after HBO treatment of volunteers. Under the same conditions, superoxide dismutase, catalase and the DNA repair enzymes apurinic endonuclease and DNA polymerase beta were not enhanced in expression. We also show that protection against the induction of DNA damage by H(2)O(2) in lymphocytes even occurs with a shortened HBO treatment which did not induce significant DNA damage by itself. Our results suggest that increased sequestration of iron as a consequence of induced HO-1 might be involved in the adaptive protection after HBO treatment and that the induction of DNA damage is not the trigger for adaptive protection.
Article
Findings in recent years strongly suggest that the stress-inducible gene heme oxygenase (HO)-1 plays an important role in protection against oxidative stress. Although the mechanism(s) by which this protection occurs is poorly understood, we hypothesized that the gaseous molecule carbon monoxide (CO), a major by-product of heme catalysis by HO-1, may provide protection against oxidative stress. We demonstrate here that animals exposed to a low concentration of CO exhibit a marked tolerance to lethal concentrations of hyperoxia in vivo. This increased survival was associated with highly significant attenuation of hyperoxia-induced lung injury as assessed by the volume of pleural effusion, protein accumulation in the airways, and histological analysis. The lungs were completely devoid of lung airway and parenchymal inflammation, fibrin deposition, and pulmonary edema in rats exposed to hyperoxia in the presence of a low concentration of CO. Furthermore, exogenous CO completely protected against hyperoxia-induced lung injury in rats in which endogenous HO enzyme activity was inhibited with tin protoporphyrin, a selective inhibitor of HO. Rats exposed to CO also exhibited a marked attenuation of hyperoxia-induced neutrophil infiltration into the airways and total lung apoptotic index. Taken together, our data demonstrate, for the first time, that CO can be therapeutic against oxidative stress such as hyperoxia and highlight possible mechanism(s) by which CO may mediate these protective effects.
Article
Rat fetal lung cells (RFL-6) were transiently transfected with a full-length rat heme oxygenase (HO)-1 cDNA construct and then exposed to hyperoxia (95% O 2 -5% CO 2 ) for 48 h. Total HO activity and HO-1 protein were measured as well as cell viability, lactate dehydrogenase (LDH) release, protein oxidation, lipid peroxidation, and total glutathione to measure oxidative injury. HO-1 overexpression resulted in increased total HO activity (2-fold), increased HO-1 protein (1.5-fold), and increased cell proliferation. Immunohistochemistry revealed perinuclear HO-1 localization, followed by migration to the nucleus by day 3. Decreased cell death, protein oxidation, and lipid peroxidation but increased LDH release and glutathione depletion were seen with HO-1 overexpression. Reactive iron content could not explain the apparent loss of cell membrane integrity. With the addition of tin mesoporphyrin, total HO activity was decreased and all changes in injury parameters were normalized to control values. We conclude that moderate overexpression of HO-1 is protective against oxidative injury, but we speculate that there is a beneficial threshold of HO-1 expression.
Article
Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. To assess the potential role of Hmox1 in cellular antioxidant defense, we analyzed the responses of cells from mice lacking functional Hmox1 to oxidative challenges. Cultured Hmox1(-/-) embryonic fibroblasts demonstrated high oxygen free radical production when exposed to hemin, hydrogen peroxide, paraquat, or cadmium chloride, and they were hypersensitive to cytotoxicity caused by hemin and hydrogen peroxide. Furthermore, young adult Hmox1(-/-) mice were vulnerable to mortality and hepatic necrosis when challenged with endotoxin. Our in vitro and in vivo results provide genetic evidence that up-regulation of Hmox1 serves as an adaptive mechanism to protect cells from oxidative damage during stress.
Article
Hyperbaric oxygen (HBO) treatment (i.e. exposure to 100% oxygen at a pressure of 2.5 ATA for a total of three 20 min periods) of human subjects caused clear and reproducible DNA effects in the comet assay with leukocytes. Interestingly, DNA damage was detected only after the first treatment and not after further treatments under the same conditions, indicating an increase in antioxidant defences. We now demonstrate that blood taken 24 h after HBO treatment is well protected against the in vitro induction of DNA damage by hydrogen peroxide (H 2 O 2 ). H 2 O 2 treatment caused a significant induction of DNA effects in the comet assay and chromosome breakage in the micronucleus test in the blood of volunteers before HBO. The same treatment did not cause genotoxic effects 24 h after HBO. This protective effect lasted for at least 1 week. Experiments with isolated lymphocytes gave similar results, indicating that the adaptive response is a cellular effect. The cells were not comparably protected against the genotoxic effects of γ-irradiation, suggesting increased scavenging of reactive oxygen species distant from nuclear DNA.
Article
Hyperbaric oxygen (HBO) treatment as used therapeutically (i.e., exposure to 100% oxygen at a pressure of 1.5 bar for a total of 60 min) has been shown to induce DNA damage in the alkaline comet assay with leukocytes from test subjects. Under these conditions, HBO did not lead to an induction of gene- and chromosome mutations. Due to known toxic effects, exposure of humans to HBO is limited and possible genetic consequences of HBO could not be completely evaluated in vivo. We thus established an in vitro HBO model, where human blood cells or V79 cells were exposed to hyperbaric oxygen (98% O2 and 2% CO2 at a pressure of either 1.5 or 3 bar) for up to 3 hr in a temperature-controlled hyperbaric chamber. Using the comet assay, we found exposure-related genotoxic effects in V79 cells, whole blood, and isolated lymphocytes. V79 cells showed the highest sensitivity toward HBO-induced DNA damage, and the exposure conditions applied to blood in vitro, to induce DNA migration, had to be higher than those used in vivo. We could also show that prolonged HBO treatment clearly increased the frequency of micronuclei in V79 cells, whereas it exerted only a marginal effect on the frequency of hprt mutations. These results demonstrate that HBO treatment of cell cultures is a well-suited model for investigating the biological significance of oxidative stress. The relationship between oxygen-induced DNA lesions and the formation of gene- and chromosome mutations is discussed. Environ. Mol. Mutagen. 34:291–296, 1999. © 1999 Wiley-Liss, Inc.
Article
Heme oxygenase the rate-limiting step in the degradation of heme to bilirubin, generates carbon monoxide. This gaseous molecule plays important roles in neuronal signaling and modulation of vascular tone. Additionally, carbon monoxide is involved in some pathological conditions (e.g., ischemia, endotoxic shock, excitotoxicity) as a protective or toxic factor. Bilirubin, another heme metabolite, exhibits intriguing biological activities as an antioxidant, an antimutagen, and an anti-complement agent. Vital functions and the dual nature displayed by these two heme metabolites are discussed.
Article
Hyperbaric oxygen (HBO) treatment as used therapeutically (i.e., exposure to 100% oxygen at a pressure of 1.5 bar for a total of 60 min) has been shown to induce DNA damage in the alkaline comet assay with leukocytes from test subjects. Under these conditions, HBO did not lead to an induction of gene- and chromosome mutations. Due to known toxic effects, exposure of humans to HBO is limited and possible genetic consequences of HBO could not be completely evaluated in vivo. We thus established an in vitro HBO model, where human blood cells or V79 cells were exposed to hyperbaric oxygen (98% O(2) and 2% CO(2) at a pressure of either 1.5 or 3 bar) for up to 3 hr in a temperature-controlled hyperbaric chamber. Using the comet assay, we found exposure-related genotoxic effects in V79 cells, whole blood, and isolated lymphocytes. V79 cells showed the highest sensitivity toward HBO-induced DNA damage, and the exposure conditions applied to blood in vitro, to induce DNA migration, had to be higher than those used in vivo. We could also show that prolonged HBO treatment clearly increased the frequency of micronuclei in V79 cells, whereas it exerted only a marginal effect on the frequency of hprt mutations. These results demonstrate that HBO treatment of cell cultures is a well-suited model for investigating the biological significance of oxidative stress. The relationship between oxygen-induced DNA lesions and the formation of gene- and chromosome mutations is discussed.
Article
Cells respond to metabolic perturbations by producing specific stress proteins. Exposure of mammalian cells to various forms of oxidative stress induces haem oxygenase, the rate-limiting enzyme in haem degradation. This response is proposed to represent an antioxidant defence operating at two different stages simultaneously. It (i) decreases the levels of the potential pro-oxidants haem and haem proteins such as cytochrome P-450 and protoporphyrinogen oxidase, and (ii) increases the tissue concentrations of antioxidatively active bile pigments.
Article
Bilirubin, the end product of heme catabolism in mammals, is generally regarded as a potentially cytotoxic, lipid-soluble waste product that needs to be excreted. However, it is here that bilirubin, at micromolar concentrations in vitro, efficiently scavenges peroxyl radicals generated chemically in either homogeneous solution or multilamellar liposomes. The antioxidant activity of bilirubin increases as the experimental concentration of oxygen is decreased from 20% (that of normal air) to 2% (physiologically relevant concentration). Furthermore, under 2% oxygen, in liposomes, bilirubin suppresses the oxidation more than alpha-tocopherol, which is regarded as the best antioxidant of lipid peroxidation. The data support the idea of a "beneficial" role for bilirubin as a physiological, chain-breaking antioxidant.
Article
Accumulating evidence suggests that oxidative stress plays a central role in the pathogenesis of many pulmonary diseases including adult respiratory distress syndrome, emphysema, asthma, bronchopulmonary dysplasia, and interstitial pulmonary fibrosis. The morbidity and mortality of these diseases remain high even with optimal medical management. In our attempts to devise new therapies for these disorders, it is crucial to improve our understanding of the basic mechanism(s) of oxidant-induced lung injury. A major line of investigation seeks to characterize the cellular and molecular responses of the lung to oxidant insults. Much progress has been made in our understanding of the role of the "classic" antioxidant enzymes (e.g., superoxide dismutase, catalase, glutathione peroxidase) in mediating the lung's resistance against oxidant lung injury. However, it is becoming clear that other oxidant-induced gene products may also play vital roles in the lung's adaptive and/or protective response to oxidative stress. One such stress-response protein is heme oxygenase-1, HO-1. Since the identification of HO-1 in 1968, many of the studies involving this enzyme were understandably focused on the regulation and function of HO-1 in heme metabolism. This emphasis is self-evident as HO-1 catalyzes the first and rate-limiting step in heme degradation. Interestingly, however, evidence accumulated over the past 25 years demonstrates that HO-1 is induced not only by the substrate heme but also by a variety of non-heme inducers such as heavy metals, endotoxin, heat shock, inflammatory cytokines, and prostaglandins. The chemical diversity of HO-1 inducers led to the speculation that HO-1, besides its role in heme degradation, may also play a vital function in maintaining cellular homeostasis. Further support for this hypothesis was provided by Tyrrell and colleagues who showed in 1989 that HO-1 is also highly induced by a variety of agents causing oxidative stress. Subsequently, many investigators have focused their attention on the function and regulation of HO-1 in various in vitro and in vivo models of oxidant-mediated cellular and tissue injury. The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. This review will focus on the current understanding of the physiological significance of HO-1 induction and the molecular regulation of HO-1 gene expression in response to oxidative stress. We hope that this discussion will stimulate interest and investigations into a field which is still largely uncharted in the pulmonary research community.
Article
The heme oxygenase (HO) system consists of two forms identified to date: the oxidative stress-inducible protein HO-1 (HSP32) and the constitutive isozyme HO-2. These proteins, which are different gene products, have little in common in primary structure, regulation, or tissue distribution. Both, however, catalyze oxidation of heme to biologically active molecules: iron, a gene regulator; biliverdin, an antioxidant; and carbon monoxide, a heme ligand. Finding the impressive heme-degrading activity of brain led to the suggestion that "HO in brain has functions aside from heme degradation" and to subsequent exploration of carbon monoxide as a promising and potentially significant messenger molecule. There is much parallelism between the biological actions and functions of the CO- and NO-generating systems; and their regulation is intimately linked. This review highlights the current information on molecular and biochemical properties of HO-1 and HO-2 and addresses the possible mechanisms for mutual regulatory interactions between the CO- and NO-generating systems.
Article
Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. To assess the potential role of Hmox1 in cellular antioxidant defense, we analyzed the responses of cells from mice lacking functional Hmox1 to oxidative challenges. Cultured Hmox1(-/-) embryonic fibroblasts demonstrated high oxygen free radical production when exposed to hemin, hydrogen peroxide, paraquat, or cadmium chloride, and they were hypersensitive to cytotoxicity caused by hemin and hydrogen peroxide. Furthermore, young adult Hmox1(-/-) mice were vulnerable to mortality and hepatic necrosis when challenged with endotoxin. Our in vitro and in vivo results provide genetic evidence that up-regulation of Hmox1 serves as an adaptive mechanism to protect cells from oxidative damage during stress.
Article
Hyperbaric oxygen (HBO) treatment as used therapeutically has been shown to induce DNA damage in the alkaline comet assay with leukocytes from test subjects. Using formamidopyrimidine-DNA glycosylase, a DNA repair enzyme which specifically nicks DNA at sites of 8-oxoguanines and formamidopyrimidines, we have detected enhanced DNA migration, indicating significant oxidative base damage, after HBO treatment. Increased DNA damage was seen immediately at the end of treatment, while 24 h later no effect was found. We now show that HBO-induced DNA strand breaks and oxidative base modifications are rapidly repaired, leading to a reduction in induced DNA effects of > 50% during the first hour. A similar decrease was found in blood taken immediately after exposure and post-incubated for 2 h at 37 degrees C in vitro and in blood taken and analysed 2 h after exposure, suggesting similar repair activities in vitro and in vivo. When the same blood samples showing increased DNA damage after HBO in the comet assay were analysed in the micronucleus test, no indications of induced chromosomal breakage in cultivated leukocytes could be obtained. The results suggest that the HBO-induced DNA effects observed with the comet assay are efficiently repaired and are not manifested as detectable chromosome damage.
Article
The gene expression of heme oxygenase-1 (HO-1) was studied in mammalian cell lines exposed to hyperoxia. Northern blot analysis demonstrated that hyperoxic exposure increased the HO-1 mRNA levels in various types of cells, including human hepatoma (HepG2) cells. This increase was time- and dose-dependent, and reversible. The HO-1 mRNA levels in HepG2 cells were increased to 2.3- and 4.2-fold of the control by hyperoxic exposure of 6 and 23 h, respectively. Cycloheximide and actinomycin D inhibited the increases in the HO-1 mRNA level produced by hyperoxia, indicating that response to hyperoxia is dependent on de novo protein synthesis and mRNA transcription. Antioxidants, desferrioxamine (DES) and o-phenanthroline (OP) partially inhibited the HO-1 mRNA elevation by hyperoxia. In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. OP, an antioxidant and a bivalent metal chelator, blocked the HO-1 mRNA elevation induced either by hyperoxia or by the three ROI generators. In contrast to OP, N-acetylcysteine (NAC), an antioxidant and membrane-permeable reducing reagent, enhanced the HO-1 mRNA elevation induced by hyperoxia, although NAC inhibited the mRNA elevation induced by NaAsO2, CdCl2 and H2O2. These results indicate that oxygen tension regulates HO-1 gene expression and suggest that hyperoxia-specific and redox-sensitive regulators may be involved in hyperoxia-mediated HO-1 gene expression.
Article
The comet assay or single-cell gel (SCG) test is becoming established as a useful technique for studying DNA damage and repair. In this microgel electrophoresis technique, a small number of cells suspended in a thin agarose gel on a microscope slide is lysed, electrophoresed, and stained with a fluorescent DNA binding dye. Cells with increased DNA damage display increased migration of chromosomal DNA from the nucleus toward the anode, which resembles the shape of a comet (Fig. 1). In its alkaline version, which is mainly used, DNA strand breaks and alkali-labile sites become apparent, and the amount of DNA migration indicates the amount of DNA damage in the cell. The comet assay combines the simplicity of biochemical techniques for detecting DNA single-strand breaks and/or alkali-labile sites with the single-cell approach typical of cytogenetic assays. The advantages of the SCG test include its simple and rapid performance, its sensitivity for detecting DNA damage, the analysis of data at the level of the individual cell, the use of extremely small samples, and its applicability to virtually any eukaryote cell population. Apart from image analysis, which greatly facilitates and enhances the possibilities of comet measurements, the cost of performing the assay is extremely low. The comet assay has already been used in many studies to assess DNA damage and repair induced by various agents in a variety of cells in vitro and in vivo (for a review, see 1,2). Fig. 1.Photomicrographs of human lymphocytes in the comet assay. (A) Untreated cell (control). (B) Cell exhibiting increased DNA migration after mutagen treatment.
Article
Hyperbaric oxygen (HBO) treatment (i.e., exposure to 100% oxygen at a pressure of 2.5 atmosphere absolute (ATA) for a total of 3 x 20 min periods) of human subjects induced DNA damage in the alkaline comet assay with leukocytes and protected against the DNA damaging effects of subsequent in vivo HBO exposures. Furthermore, blood taken 24 h after the first HBO was well protected against the in vitro induction of genotoxic effects by hydrogen peroxide. To investigate the mechanisms which led to this apparent adaptive response, we determined the antioxidant status of blood from subjects before and after HBO. We did not find differences in the plasma concentrations of the antioxidant vitamins A, C and E after HBO treatment. HBO had also no effect on the 'antioxidant power' of the plasma as measured with the FRAP-assay or on the concentration of reduced glutathione determined in the plasma or in lymphocytes. Red cell concentrate activities of superoxide dismutase, catalase, glutathione peroxidase were not influenced by HBO. In contrast, synthesis of the heat shock protein HSP70 which has been implicated to play an important role in cellular protection against oxidative stress, was significantly induced in lymphocytes after a single HBO treatment. To investigate whether intake of antioxidants may protect against HBO-induced DNA damage, we supplemented subjects with vitamin E (800 mg for 7 days) or with N-acetylcysteine (400 mg, 1 h before the HBO treatment). However, these supplementations did not influence the induction of DNA damage by HBO.
Article
The HO-1 isoenzyme is an early stress response gene regulated by many forms of oxidative stress. The HO-2 isoenzyme is predominantly a constitutive enzyme, which may serve to sequester heme as well as degrade it. All HO enzyme activity results in the degradation of heme and the production of antioxidant bile pigments, which would favor an antioxidant role for the enzyme. In fact, in oxidative stress in vitro, HO-1 is protective (91-94) but within a narrow threshold of overexpression (93,94) in some models, since iron released in the HO reaction may obviate any cytoprotective effect (Fig. 3). So far, HO-2 appears to be beneficial in oxygen toxicity in vivo, but the consequences of HO-2 overexpression have not yet been tested. It will be important to better define the role of each HO isoenzyme in oxidative stress so as to determine whether enhancing these complex systems could alleviate some of the cellular changes seen as a result of oxidative injury. Furthermore, prior to considering therapeutic maneuvers to enhance HO, a complete understanding of the physiologic consequences of HO-1 induction and associated reactions, in each particular setting, will be crucial.
Article
We recently showed that treatment of V79 cells with hyperbaric oxygen (HBO) efficiently induced DNA effects in the comet assay and chromosomal damage in the micronucleus test (MNT), but did not lead to gene mutations at the hprt locus. Using the comet assay in conjunction with bacterial formamidopyrimidine DNA glycosylase (FPG protein), we now provide indirect evidence that the same treatment leads to the induction of 8-oxoguanine, a premutagenic oxidative DNA base modification in V79 and mouse lymphoma (L5178Y) cells. We also demonstrate that HBO efficiently induces mutations in the mouse lymphoma assay (MLA). Exposure of L5178Y cells to HBO (98% O(2); 3bar) for 2h caused a clear mutagenic effect in the MLA, which was further enhanced after a 3h exposure. As this mutagenic effect was solely due to the strong increase of small colony (SC) mutants, we suggest that HBO causes mutations by induction of chromosomal alterations. Molecular characterization of induced SC mutants by loss of heterozygosity (LOH) analysis showed an extensive loss of functional tk sequences similar to the pattern found in spontaneous SC mutants. This finding confirmed that the majority of HBO-induced mutants is actually produced by a clastogenic mechanism. The induction of point mutations as a consequence of induced oxidative DNA base damage seems to be of minor importance.
Article
Since 1922 when Wu proposed the use of the Folin phenol reagent for the measurement of proteins (l), a number of modified analytical pro- cedures ut.ilizing this reagent have been reported for the determination of proteins in serum (2-G), in antigen-antibody precipitates (7-9), and in insulin (10). Although the reagent would seem to be recommended by its great sen- sitivity and the simplicity of procedure possible with its use, it has not found great favor for general biochemical purposes. In the belief that this reagent, nevertheless, has considerable merit for certain application, but that its peculiarities and limitations need to be understood for its fullest exploitation, it has been studied with regard t.o effects of variations in pH, time of reaction, and concentration of react- ants, permissible levels of reagents commonly used in handling proteins, and interfering subst.ances. Procedures are described for measuring pro- tein in solution or after precipitation wit,h acids or other agents, and for the determination of as little as 0.2 y of protein.
Regulation and role of heme oxygenase in oxidative injury
  • P Dennery
Dennery,P. (2000) Regulation and role of heme oxygenase in oxidative injury. Curr. Top. Cell. Regul., 36, 181–199.
The heme oxygenase system: a regulator of second messenger gases
  • M Maines
Maines,M. (1997) The heme oxygenase system: a regulator of second messenger gases. Annu. Rev. Pharmacol. Toxicol., 37, 517-554.