Next Article in Journal
GNAi2/gip2-Regulated Transcriptome and Its Therapeutic Significance in Ovarian Cancer
Next Article in Special Issue
Hyperbaric Oxygen Treatment—From Mechanisms to Cognitive Improvement
Previous Article in Journal
Histatin 5 Metallopeptides and Their Potential against Candida albicans Pathogenicity and Drug Resistance
Previous Article in Special Issue
Molecular Biomarkers of Oxygen Therapy in Patients with Diabetic Foot Ulcers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Effects of Hyperbaric Oxygenation on Oxidative Stress, Inflammation and Angiogenesis

by
Silke D. De Wolde
1,2,*,
Rick H. Hulskes
1,3,
Robert P. Weenink
1,2,
Markus W. Hollmann
1 and
Robert A. Van Hulst
1,2
1
Department of Anesthesiology, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands
2
Department of Hyperbaric Medicine, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands
3
Department of Surgery, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Biomolecules 2021, 11(8), 1210; https://doi.org/10.3390/biom11081210
Submission received: 3 July 2021 / Revised: 30 July 2021 / Accepted: 9 August 2021 / Published: 14 August 2021
(This article belongs to the Special Issue Oxygen Therapy)

Abstract

:
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.

1. Introduction

Since the adjunctive use of hyperbaric oxygen therapy (HBOT) was first described in 1879 [1], it has been further explored and is nowadays a widely accepted treatment in several diseases, such as delayed radiation injury, diabetic foot ulcers, carbon monoxide poisoning, decompression sickness and arterial gas embolism [2]. The Undersea and Hyperbaric Medical Society (UHMS) describes HBOT as an intervention whereby patients breathe near 100% oxygen while being pressurized to at least 1.4 atmosphere absolute (ATA) in a hyperbaric chamber [1]. Currently, the UHMS has accepted 14 indications for HBOT [3], yet new applications of HBOT have been described, including preconditioning for surgery [4,5,6,7].
Several cohort studies and randomized controlled trials, executed in different surgical procedures (e.g., abdominoplasty and pancreaticoduodenectomy), reported lower postoperative complication rates and a reduced length of stay on the intensive care unit after preoperative HBOT [4,5,6,7]. As the occurrence of postoperative complications is associated with worse short-term and long-term outcomes [8], a decrease in psychosocial well-being [9] and higher healthcare costs [10], HBOT may prevent those adverse effects of surgery.
To realize this perioperative protective effect, HBOT must be able to prevent infection and increase wound healing. It is likely that oxidative stress, which has been confirmed to be the main effect of HBOT [11], plays an activating role in the mechanisms underlying the therapeutic pathway of preconditioning for surgery with HBOT. An increase in reactive oxygen species (ROS) levels is associated with enhanced pathogen clearance [12]. Furthermore, ROS induce the synthesis of several growth factors, such as vascular endothelial growth factor (VEGF), placental growth factor (PGF) and angiopoietin (Ang) 1 and 2 and recruit stem cells from the bone marrow, which are responsible for neovascularization [13]. However, a frequently mentioned argument against the use of HBOT revolves around the induction of oxidative stress as well, since higher levels of ROS and reactive nitrogen species (RNS) may lead to oxidative and nitrosative damage, mitochondrial aging, genotoxicity and maintenance of (chronic) inflammation [14,15,16].
The aim of this review is to gain more insight into the mechanisms of HBOT by assessing its effect on oxidative stress, inflammation and angiogenesis markers in humans. More insight into these effects of HBOT will predict and underpin the outcome of innovative uses of HBOT and balance its benefits against potential damage. No systematic overview of research into these parameters in human beings has yet been published.

2. Methods

A search of the literature was performed in MEDLINE and EMBASE on 2 November 2020. Key terms used in the search were ‘hyperbaric oxygen’ and ‘oxidative stress’, ‘inflammation’, or ‘wound healing’. The results were not restricted as no filters were applied. The detailed literature search can be found in Appendix A (see Table A1 and Table A2).
All studies found were screened on title and abstract by one reviewer (S.D.D.W.), who excluded those studies that met any of the following criteria: (1) absence of abstract, (2) congress abstract, errata or guideline, (3) case report (defined as five or less patients), (4) narrative review, (5) animal research, (6) no treatment with HBOT, or (7) one of the following outcome measures: cure, complication rate, or a disease-specific outcome parameter. The same reviewer assessed the full-text of the remaining studies. The following inclusion criteria were applied: (1) measurement of at least one marker of oxidative stress, inflammation, or angiogenesis before and after HBOT, (2) study in humans (or human material) and (3) English full-text available. EndNote X9 was used to keep track of the screening process.
The included studies were divided into an “in vivo” and “in vitro” group. In vivo studies were performed in a clinical setting in which all subjects were at least pressurized once, whereas in vitro studies obtained human material what was subsequently exposed to HBOT. Information on first author, publication year, investigated parameters and patient (in vivo)/sample (in vitro) characteristics and results (solely of the parameters of interest) were extracted. Outcomes of statistical tests with a p-value < 0.05 were considered significant. All information was extracted by hand and documented in Microsoft Excel (v16.0).

3. Results

3.1. Eligible Studies

The search retrieved 9618 records. After removing duplicates and screening of title and abstract, 216 studies were screened full-text. Finally, 137 articles were included in this review (see Figure 1). Most of the included articles were clinical studies (n = 98) and performed in patients with diabetes mellitus and/or non-healing chronic wounds (n = 27). Furthermore, 27 articles describing the effect of HBOT in healthy volunteers (including divers) were found. Sixteen included studies reported on other biomarkers than described in Table 1, Table 2 and Table 3 (data not shown) [17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32].

3.2. Oxidative Stress

In total, 74 articles reporting on the effect of HBOT on oxidative stress were found. Subjects mainly received one session of HBOT in a hyperbaric chamber pressurized to 2–2.5 ATA (203–253 kPa), yet in seven studies a wet exposure to hyperbaric oxygen (i.e., a dive) to up to 6 ATA (608 kPa) was employed. Nearly 40% (n = 21) of the clinical studies were conducted in healthy volunteers (see Table A3). Catalase, glutathione peroxidase (GPx), malondialdehyde (MDA), nitric oxide synthase (NOS), ROS, RNS, superoxide dismutase (SOD) and thiobarbituric acid reactive substances (TBARS) were the most frequent markers of interest (see Table 1).
A clear stimulating effect of HBOT on ROS (see Table 1) was found. Nonetheless, two out of the three studies assessing hydrogen peroxide described lower concentrations after HBOT [33,42] (see Table A3). NOS and RNS concentrations seem to increase after HBOT as well, although this effect was less pronounced, which can be explained by a repeatedly reported decrease in exhaled nitric oxygen [61,69,70]. Timing of sampling may also play a role, as several articles only noted an increase in inducible NOS or nitrite three hours after the end of an HBOT session [34,49,55].
Not only the presence of NOS, RNS and ROS has been investigated, but also their effects on lipids, proteins, carbohydrates and DNA/RNA (see Table 1). Little research has been done regarding protein and carbohydrate modifications following HBOT, but no effect or a stimulating effect on lipid peroxidation, resulting in MDA and other aldehydes (TBARS), has been reported in various studies. DNA-damaging effects of HBOT were not demonstrated employing the most commonly used DNA-lesion-marker 8-hydroxydeoxyguanosine [146].
Concerning the concentrations of anti-oxidative enzymes that protect against the potentially harmful effects of oxidative stress, such as catalase, SOD and GPx, conflicting results were found (see Table 1). In general, no effect or an indication for an increasing effect of HBOT on the enzyme activity of those antioxidants has been demonstrated. HBOT may have a uniform effect on SOD and catalase, as most of the studies reported increased, decreased, or stable SOD and catalase levels and, thus, no differences in effect of HBOT between these two enzymes [76,80,81,83,85,94,97,100]. However, a difference between SOD and/or catalase concentrations in respectively plasma and erythrocytes has been reported [55,62,63]. Benedetti et al. [80] and Dennog et al. [94] describe no effect of HBOT on the free radical trapping anti-oxidants with an exogenous origin, such as vitamin A, vitamin C and vitamin E [149].

3.3. Inflammation

Of the 140 studies included, 58 articles describing inflammatory markers were identified. Most of the research included at least three HBOT-sessions, yet study protocols consisting of 20–40 sessions were common, in particular in articles reporting acute-phase proteins (see Table A4). Popular variables of interest were interleukins (IL) (n = 31), acute-phase proteins (n = 26) and tumor necrosis factor-α (TNF-α) (n = 25) (see Table 2).
Concerning acute phase proteins, a decreasing effect of HBOT on (high-sensitivity) C-reactive protein ((hs-)CRP) was found as 75% (n = 12) of the studies investigating (hs-) CRP reported lower concentrations post-HBOT. Strikingly, HBOT may have a stimulating impact on granulocyte-colony stimulating factor and an inhibiting effect on insulin-like growth factor-1, both reflecting a pro-inflammatory state [150] (see Table 2).
No impact of HBOT on most interleukin concentrations (IL-2, IL-3, IL-5, IL-7, IL-9, IL-12p70, IL-13, IL-15, IL-17, IL-18 and IL-22) has been demonstrated, although Hao et al. [111] reported a decrease in IL-12p40 levels (see Table A4). Concerning the proinflammatory interleukins, a potentially inhibiting effect of HBOT on IL-1β, IL-6 and IL-8 was found, whereas Dhadmodharan et al. [45] suggested an increase in IL-1α levels. On the other hand, a rise in the anti-inflammatory IL-1Ra was reported, alongside a possible inhibiting effect of HBOT on IL-10 and no effect on IL-4. Both results support an anti-inflammatory state (see Table 2) [151].
In line with the outcomes regarding (hs-)CRP and interleukins, an anti-inflammatory effect of HBOT was also shown by decreasing levels of the pro-inflammatory cytokines interferon-γ (IFN-γ), nuclear factor kappa B (NF-κB) and TNF-α (see Table 2). However, HBOT may have an initial pro-inflammatory effect, as some studies described an increase in TNF-α during or shortly after HBOT [87,127,134].

3.4. Angiogenesis

Concerning the angiogenesis research, 34 studies were found in addition to the earlier mentioned studies reporting on interleukins, interferons, insulin-like growth factor 1 (IGF-1), NF- κB and TNF-α. Most of the articles described angiogenesis-inducing cytokines or growth factors and were performed in clinical setting (n = 20). However, five out of seven studies on downstream effectors of angiogenesis were conducted in vitro (see Table A5). Epidermal growth factor (EGF), extracellular signal-regulated kinase (ERK), (basic) fibroblast growth factor, tumor growth factor-β (TGF-β), VEGF, IFN-γ, IL-6, IL-8, NF-κB and TNF-α (see Table 2 and Table 3) were the only angiogenesis markers reported in at least five articles.
HBOT most likely has a stimulating effect on various growth factors involved in angiogenesis (i.e., EGF, hematopoietic growth factor, keratinocyte growth factor, PGF and VEGF). This effect may only be present shortly after the intervention, since several studies with repeated HBOT sessions described no differences in pre-HBOT values or only a raise after the first session (and not after following sessions) [62,141,147] (see Table A5). Whereas for some angiogenesis-stimulating cytokines, such as stromal cell-derived factor-1α, a similar increasing effect of HBOT was found, no or an inhibiting effect on TGF was seen. HBOT seems not to affect the cytokine receptors (see Table 3).
HBOT decreased matrix metalloproteinases (MMPs) [52,54,72]. According to Niu et al. [52,72], the effect on MMPs is delayed and only manifests after two or three HBOT sessions. Hypoxia-inducible factor-1α (HIF-1α) and NF-κB were inhibited by HBOT (see Table 2 and Table 3), although Anguinano-Hernandez et al. [125] described an increase in NF-κB in the cytosol.
As HBOT causes an increase in angiogenesis-promoting growth factors and cytokines, one would also expect a stimulating effect on the downstream effectors of blood vessel formation. However, inconsistent outcomes were reported (see Table 3). The phosphatidylinositol-3 kinase (PI3K)/AKT pathway was upregulated and the ERK and p38 mitogen-activated protein kinase (p38 MAPK) pathways were downregulated. Therefore, HBOT effects on downstream effectors of blood vessel formation seem to differ depending on the intracellular effector route.

4. Discussion

This review is the first to systematically summarize the effect of HBOT on oxidative stress, inflammation and angiogenesis markers in human beings. HBOT increases the levels of oxygen radicals, which induce oxidative stress. An anti-inflammatory action of HBOT was demonstrated by decreasing concentrations of several pro-inflammatory markers. Furthermore, HBOT seems to stimulate the release of angiogenesis-promoting cytokines, including growth factors.
In the light of previous research, reporting a link between oxidative stress and a pro-inflammatory state [152,153,154], it is remarkable that HBOT leads to a more anti-inflammatory state. However, these findings do correspond with studies into the effects of HBOT using thermal imaging, in which a decrease in wound temperature was found [155,156]. This temperature reduction could indicate a local decline in inflammation. This anti-inflammatory effect is likely mediated by the inhibition of NF-κB, a transcription factor for pro-inflammatory genes [157,158,159]. A direct anti-inflammatory action of HBOT seems less probable, since no differences in the concentrations of anti-inflammatory markers (except IL-1Ra) were noted. Although beyond the scope of this review, Yu et al. [160] have shown in an animal model that HBOT decreases the NF-κB concentrations by higher release of IκBα, which is an inhibitor of NF-κB and degrades under hypoxic circumstances [161]. An increase in IκBα along with a decrease in NF-κB after HBOT was also seen in the only study in the current review reporting on IκBα [52]. Therefore, hyperoxia generated during HBOT may stimulates the preservation of IκBα and thereby inhibits NF-κB release, resulting in less gene transcription of pro-inflammatory cytokines and, thus, an anti-inflammatory state despite oxidative stress.
NF-κB is not only a crucial transcription factor in inflammation, but also plays a role, together with HIF-1α, in the induction of angiogenesis. Growth factors and other angiogenesis-promoting cytokines induce new vessel formation by increased expression of pro-angiogenesis genes, which is mediated by NF-κB or (under hypoxia) HIF-1α [162,163]. Since the current review demonstrates an inhibiting effect of HBOT on both transcription factors and little research, with contradicting outcomes, into the downstream effectors of angiogenesis (i.e., PI3K, Akt, p38 MAPK, ERK) has been done, it is unclear how increased levels of pro-angiogenesis growth factors and cytokines actually induce increased tube formation, as shown by Anguiano-Hernandez et al. [125], Lin et al. [130] and Shyu et al. [40]. Thus, further research into the relation between NF-κB, HBOT and the angiogenesis pathways is needed.
Another striking finding concerning angiogenesis is that several articles reported an increase in growth factors only or particularly after the first HBOT session [40,141,147], while it is common to conduct 20–40 sessions for chronic non-healing wounds or radiation-induced tissue injury (indications strongly relying on the angiogenesis effects of HBOT) [2]. Furthermore, Sureda et al. [62] describe, in the only in vivo study assessing the effect of HBOT on growth factors at several time points during follow-up, an increase in VEGF immediately after each session, yet VEGF levels determined pre-session #5 and #20 were similar to the baseline (pre-session #1) value. Those findings possibly suggest a short pro-angiogenesis effect of HBOT. However, due to a shortage of studies reporting on angiogenesis markers on a daily or weekly basis during a treatment protocol including 20–40 sessions, it remains unclear which markers are involved in this short-term effect of HBOT and whether other factors play a role in this angiogenesis process.
The aim of this review was to gather a comprehensive overview of the effects of HBOT on oxidative stress, inflammation and angiogenesis. We must conclude that existing research does not allow for a complete understanding of the physiology underlying new promising treatment modalities for HBOT, such as preconditioning for surgery. Due to the heterogeneity of included patient populations and the inclusion of studies in healthy volunteers, it is difficult to extrapolate findings to the surgical patient in general. Furthermore, this review did not focus on clinical outcomes related to inflammation, angiogenesis and oxidative stress, making it impossible to determine the implications of the described findings in practice. In conclusion, hyperoxia and oxidative stress induced by HBOT affect inflammation and angiogenesis markers, but whether hyperoxia and oxidative stress induce a clinically relevant decrease in inflammation and increase in angiogenesis remains unclear and needs to be further investigated before innovative interventions can be widely applied.

Author Contributions

Conceptualization, R.P.W. and R.A.V.H.; methodology, S.D.D.W., R.P.W. and R.A.V.H.; validation, S.D.D.W., R.H.H., R.P.W., M.W.H. and R.A.V.H.; formal analysis, S.D.D.W.; investigation, S.D.D.W.; data curation, S.D.D.W.; writing—original draft preparation, S.D.D.W.; writing—review and editing, S.D.D.W., R.H.H., R.P.W., M.W.H. and R.A.V.H.; visualization, S.D.D.W.; supervision, R.P.W., M.W.H. and R.A.V.H.; project administration, R.P.W. and R.A.V.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Search strategy used in PubMed (MEDLINE).
Table A1. Search strategy used in PubMed (MEDLINE).
Key TermsMesh-TermsTitle/Abstract-Terms
Hyperbaric oxygenhyperbaric oxygenation[MeSH]hyperbaric oxygen[tiab] OR hyperbaric oxygenation[tiab] OR hyperbaric oxygen therapy[tiab] OR hyperbaric oxygen therapies[tiab] OR HBO[tiab] OR HBOT[tiab] OR hyperbaric medicine[tiab]
AND
Inflammationinflammation[MeSH] OR inflammation mediators[MeSH] OR autacoids[MeSH] OR chemokines[MeSH] OR synthetic prostaglandins[MeSH] OR interleukin[MeSH] OR infection[MeSH]inflammation[tiab] OR inflammations[tiab] OR inflammatory[tiab] OR inflammatory respons[tiab] OR inflammation mediators[tiab] OR mediators of inflammation[tiab] OR chemokines[tiab] OR slow reacting substances[tiab] OR chemotactic cytokines[tiab] OR synthetic prostaglandins[tiab] OR intercrines[tiab] OR PG analogs[tiab] OR prostaglandin analogues[tiab] OR prostaglandin analogs[tiab] OR interleukin[tiab] OR infection[tiab] OR infection and infestation[tiab] OR infections and infestations[tiab] OR infestation and infection[tiab] OR infestations and infections[tiab]
OR
Wound healingwound healing[MeSH] OR re-epithelialization[MeSH] OR angiogenesis modulating agents[MeSH] OR neovascularization, physiologic[MeSH] OR cell proliferation[MeSH]woundhealing[tiab] OR wound healing[tiab] OR cicatrization[tiab] OR re-epithelialization[tiab] OR wound epithelialization[tiab] OR angiogenesis[tiab] OR angiogenesis modulating agents[tiab] OR vasculogenesis[tiab] OR blood vessel formation[tiab] OR bloodvessel formation[tiab] OR neovascularization[tiab] OR neovascularisation[tiab] OR cell proliferation[tiab] OR endothelial proliferation[tiab] OR vascular proliferation[tiab]
OR
Oxidative stressOxidative stress[MeSH] OR nitrosative stress[MeSH] OR reactive oxygen species[MeSH] OR reactive nitrogen species[MeSH]oxidative stress[tiab] OR nitrosative stress[tiab] OR reactive oxygen species[tiab] OR reactive nitrogen species[tiab] OR peroxide[tiab] OR peroxides[tiab] OR superoxide[tiab] OR superoxides[tiab] OR hydroxy radical[tiab] OR hydroxy radicals[tiab] OR singlet oxygen[tiab] OR alpha-oxygen[tiab] OR nitric oxide[tiab] OR peroxynitrite[tiab] OR nitrogen dioxide[tiab] OR oxidant stress[tiab] OR reactive oxygen metabolite[tiab] OR reactive oxygen metabolites[tiab] OR reactive nitrogen metabolite[tiab] OR reactive nitrogen metabolites[tiab]
Table A2. Search strategy used in Ovid (EMBASE).
Table A2. Search strategy used in Ovid (EMBASE).
Key-Terms.Emtree-TermsTitle/Abstract/Author Keywords-Terms
Hyperbaric oxygenhyperbaric oxygen/ OR hyperbaric oxygen therapy/hyperbaric oxygen therapy.ti,ab,kw OR hyperbaric oxygen therapies.ti,ab,kw OR HBOT.ti,ab,kw OR hyperbaric oxygenation.ti,ab,kw OR hyperbaric oxygen.ti,ab,kw OR HBO.ti,ab,kw OR hyperbaric medicine.ti,ab,kw
AND
InflammationInflammation/ OR inflammation autocoid/ OR chemokine/ OR chronic inflammation/ OR inflammation/ OR cytokine/ OR infection/inflammation.ti,ab,kw OR inflammations.ti,ab,kw OR inflammatory.ti,ab,kw OR inflammatory respons.ti,ab,kw OR inflammation mediators.ti,ab,kw OR mediators of inflammation.ti,ab,kw OR chemokines.ti,ab,kw OR slow reacting substances.ti,ab,kw OR chemotactic cytokines.ti,ab,kw OR synthetic prostaglandins.ti,ab,kw OR intercrines.ti,ab,kw OR PG analogs.ti,ab,kw OR prostaglandin analogues.ti,ab,kw OR prostaglandin analogs.ti,ab,kw OR interleukin.ti,ab,kw OR infection.ti,ab,kw OR (infection and infestation).ti,ab,kw OR (infections and infestations).ti,ab,kw OR (infestation and infection).ti,ab,kw OR (infestations and infections).ti,ab,kw
OR
Wound healingwound healing/ OR epithelialization/ OR angiogenesis/ OR cell proliferation/woundhealing.ti,ab,kw OR wound healing.ti,ab,kw OR cicatrization.ti,ab,kw OR re-epithelialization.ti,ab,kw OR wound epithelialization.ti,ab,kw OR angiogenesis.ti,ab,kw OR neovascularization.ti,ab,kw OR neovascularisation.ti,ab,kw OR (angiogenesis modulating agents).ti,ab,kw OR vasculogenesis.ti,ab,kw OR blood vessel formation.ti,ab,kw OR bloodvessel formation.ti,ab,kw OR cell proliferation.ti,ab,kw OR endothelial proliferation.ti,ab,kw OR vascular proliferation.ti,ab,kw
OR
Oxidative stressoxidative stress/ OR nitrosative stress/ OR reactive oxygen metabolite/ OR reactive nitrogen species/oxidative stress.ti,ab,kw OR nitrosative stress.ti,ab,kw OR reactive oxygen species.ti,ab,kw OR reactive nitrogen species.ti,ab,kw OR peroxide.ti,ab,kw OR peroxides.ti,ab,kw OR superoxide.ti,ab,kw OR superoxides.ti,ab,kw OR hydroxy radical.ti,ab,kw OR hydroxy radicals.ti,ab,kw OR singlet oxygen.ti,ab,kw OR alpha-oxygen.ti,ab,kw OR nitric oxide.ti,ab,kw OR peroxynitrite.ti,ab,kw OR nitrogen dioxide.ti,ab,kw OR oxidant stress.ti,ab,kw OR reactive oxygen metabolite.ti,ab,kw OR reactive oxygen metabolites.ti,ab,kw OR reactive nitrogen metabolite.ti,ab,kw OR reactive nitrogen metabolites.ti,ab,kw
Table A3. Retrieved results on oxidative stress markers in detail.
Table A3. Retrieved results on oxidative stress markers in detail.
StudyYearDesignSubjectsMethodsType oxidative Stress MarkerOutcomeRemarks
Amount of SessionsMaximum Pressure (ATA) aMoment of Sample Taking b
Ansari et al. [100]1986In vivo18 patients; multiple sclerosis202Within 30 min post-HBOTCatalase, GPx, SODCatalase: increase
SOD: increase
GPx: no significant differences were found
No effect of HBOT was seen in the group breathing chamber air at 2 ATA
Bearden et al. [86]1999In vivo10 divers11.4–1.5 (dive)Within 15 min post-diveSOD, TBARSSOD: decrease
TBARS: increase
Benedetti et al. [80]2004In vivo12 patients; several pathological conditions152.5Immediately post-session #1 and #15Catalase, glutathione, GPx, MDA, reactive oxygen metabolites, retinol (vitamin A), SOD, α-tocopherol (vitamin E)Catalase: decrease
MDA: increase
Reactive oxygen metabolites: increase
SOD: decrease
Glutathione, GPx, retinol (vitamin A), a-tocopherol (vitamin E): no significant differences were found
Reported outcomes are based on a comparison of the pre-session values of sessions #1 and #15. No significant differences in post-session values were found
Bosco et al. [35]2018In vivo23 patients; unilateral femoral head necrosis602.5Post-session #15, #30, and #60 and pre-session #31ROSIncrease at post-session #15, post-session #30 and pre-session #31
Boykin et al. [57]2007In vivo6 patients; chronic non-healing wound202Post-session #10, post-session #20, 1 week post-HBOT, and 4 weeks post-HBOTNitric oxygenIncrease at 1 week post-HBOT and 4 weeks post-HBOT
Burgos et al. [91]2016In vivo12 young soccer players152Pre- and post-session #5, #10, and #15Antioxidant capacity, lipid hydroperoxides, uric acidNo significant differences were foundResults are possible influenced by exercising during HBOT
Chang et al. [102]2020In vivo10 healthy male volunteers12.830 min, 2 days, and 1 week post-HBOTUric acidNo significant differences were found
Chen et al. [36]2011In vitroBlood samples of healthy males11.5, 2, and 2.5?Lipid peroxides, superoxide-ionLipid peroxides: increase
Superoxide-ion: increase
Chen et al. [88]2018In vivo50 patients; acute non-cardioembolic stroke12.51 month post-HBOTTBARs, thiolsNo significant differences were found
Chen et al. [67]2007In vivo31 patients with diabetes mellitus type 2 and 29 healthy volunteers32.5Immediately post-session #1 and post-session #3Nitric oxygenDecrease in diabetes mellitus group
Cheung et al. [37]2018In vitroUmbilical cord blood enriched with CD34-cells12.524 h post-HBOTROSIncrease in nucleus and mitochondriaNo significant differences were found in the cytoplasm
Corcoran et al. [78]2017In vivo12 patients; osteonecrosis secondary to radiation therapy12.4During HBOT, immediately post-HBOT, and 30 min post-HBOTIsofurans, isoprostanesNo significant differences were found
Dejmek et al. [164]2018In vitroHuman fetal lung fibroblasts53?SODIncrease
Dennog et al. [94]1999In vivoHealthy volunteers12.5Immediately and 24 h post-HBOTCatalase, glutathione, GPx, SOD, tail moment, total antioxidant capacity, vitamin A, vitamin C, vitamin ETail moment: increase
Catalase, glutathione, GPx, SOD, total antioxidant capacity, vitamin A, vitamin C, vitamin E: no significant differences were found
Dhamodharan et al. [45]2019In vivo37 patients; diabetic foot ulcer252.220 days after the first HBOT-sessionCatalase, endothelial NOS, heme oxygenase-1, NAD(P)H dehydrogenase [quinone] 1, nitrite, nuclear factor erythroid-2 related factor 2Catalase: increase
Endothelial NOS: increase
Heme oxygenase-1: increase
NAD(P)H dehydrogenase [quinone] 1: increase
Nitrite: increase
Nuclear factor erythroid-2 related factor 2: increase
Dise et al. [92]1987In vivoAdult male volunteers13Within 60 min post-HBOT and 24 h post-HBOTglutathione, lipid hydroperoxidesGlutathione: increase
Lipid hydroperoxides: decrease
Dragic et al. [65]2020In vivo64 patients; peripherial arterial disease102.2Post-session #10Nitric oxygenNo significant differences were found
Eken et al. [14]2005In vivo15 patients202.5Immediately post-session #1, #10, and #20GPx, MDA, sister chromatide exchange, SODSister chromatide exchange: increase
GPx, MAD, SOD: no significant differences were found
Ferrer et al. [34]2007In vivo7 male divers and 12 male physically active volunteers15 (dive)/2.2Immediately and 3 h post-dive/30 min post-HBOTcatalase, GPx, hydrogen peroxide, inducible NOS, MDA, myeloperoxidase, nitriteCatalase: increase 3 h post-dive
GPx: increase
Hydrogen peroxide: increase 3 h post-dive and post-HBOT
Inducible NOS: increase 3 h post-dive
Myeloperoxidase: decrease 3 h post-dive
Nitrite: increase 3 h post-dive
MDA: no significant differences were found
Gasier et al. [63]2013In vivo12 healthy male divers31.5/215 min, 1 h, and 2 h after each sessionCatalase, GPx, nitrite, SOD, TBARSCatalase: decrease in erythrocytes post-HBOT at 2 ATA and 1 h post-HBOT at 1.5 ATA
GPx: increase in erythrocytes post-HBOT at 1.5 ATA and a decrease in erythrocytes post-HBOT at 2 ATA
TBARS: increase in erythrocytes 15 min post-HBOT at 1.5 ATA
Nitrite, SOD: no significant differences were found
No significant differences in catalase, GPX, and TBARS were found in the plasma
Grimberg-Peters et al. [44]2016In vitroNeutrophils from severely injured patients and healthy volunteers12?ROSDecreaseAfter 3 h stimulation with PMA
Gröger et al. [38]2009In vitroLymphocytes from combat swimmers, divers, and nondiving volunteers14Immediately, 1 h, and 2 h post-HBOTSuperoxide-ion, tail momentSuperoxide-ion: increase
Tail moment: increase immediately post-HBOT
No increase in superoxide radical was seen in the combat swimmers group, which had high baseline superoxide radical levels. Superoxide radical has been measured only once (at which measurement point is unknown)
Gronow et al. [74]2005In vivo28 divers and 10 volunteers11.7 (dive)/2.8?Hydrobenzoates, TBARSHydrobenzoates: increase
TBARS: increase
No significant differences were found concerning monohydrobenzoates
Gurdol et al. [68]2010In vivo18 patients; diabetic foot ulcers25/302.4Post session #25/#30Nitric oxygenNo significant differences were foundA decrease in NO levels post-HBOT was seen in the group with <50% wound healing, which had significantly higher baseline NO values
Gürdöl et al. [77]2008In vivo20 patients; type 2 diabetic with foot ulcers152.430 min post-session #1 and #15Advanced oxidation proteins products, isoprostanes, MDAAdvanced oxidation proteins products: decrease at pre-session #15
Isoprostanes: increase post-session #15
MDA: increase post-session #1
Handy et al. [99]2005In vivo31 patients; non-healing wounds202.2Immediately post-session #1 and #20Total antioxidant capacityNo significant differences were found
Kähler et al. [75]2013In vivo118 volunteers12.4/2.8?Dihydroxylated benzoateIncreaseAdministration of 100% oxygen significantly increased the dihydroxylated benzoate levels, yet pressurization had no extra effect.
Karadurmus et al. [103]2010In vivo28 patients; diabetic foot ulcers302.4Post-session #10, #20, and #30Uric acidDecrease
Kendall et al. [46]2012In vitroHuman umbilical vein endothelial cells12.4Immediately, 5 h, and 22.5 h post-HBOTEndothelial NOS, nitrate + nitrite, nitric oxygenEndothelial NOS: increase
Nitrate + nitrite, nitric oxygen: no significant differences were found
Kendall et al. [42]2013In vitroHuman umbilical vein endothelial cells12.4?Hydrogen peroxide, superoxide-ionHydrogen peroxide: decrease
Superoxide-ion: no significant differences were found
Kot et al. [93]2003In vivo96 healthy volunteers12.8Immediately post-HBOTProtein carbonyls, total antioxidant status, total thiolProtein carbonyls: increase
Total thiol: decrease
Total antioxidant status: no significant differences were found
Kozakiewicz et al. [56]2018In vivo42 healthy volunteers13?Carbonyl group, MDA, nitrate/nitrite, SODCarbonylgroep: increase
MDA: increase
Nitrate/nitrite: increase
SOD: increase
The baseline values were significantly higher (carbonyl group) and lower (nitrate/nitrite and SOD-1) in the HBOT-group compared to the control group
Lambrechts et al. [64]2013In vivo10 military divers14 (dive)/1.71 h post-dive/1 h post-HBOTNitrotyrosine, nitric oxygenNo significant differences were found
Li et al. [84]2017In vivo78 patients; chronic diabetic woundsBy average 482.430 days after the first HBOT-sessionMDA, SODMDA: decrease
SOD: increase
Li et al. [58]2018In vivo115 patients; coronary artery disease with drug-eluting stents242?Nitric oxygenIncrease
Li et al. [59]2019In vivo115 patients; coronary artery disease with coronary stent implantation242?Nitric oxygenIncrease
Li et al. [60]2018In vivo98 patients; slow coronary flow242?Nitric oxygenIncrease
Lin et al. [39]2008In vitroDetroit 551 normal human dermal fibroblasts32.5?ROSIncrease
Ma et al. [81]2013In vivo36 patients; diabetic foot ulcers202.57 and 14 days after the first HBOT-sessionCatalase, MDA, SODCatalase: increase post-session #14
MDA: increase post-session #14
SOD: increase post-session #14
Matzi et al. [82]2015In vivo23 healthy volunteers12.2During HBOT and immediately post-HBOT8-hydroxy-deoxyguanosine, GPx, MDA, oxidized low-density lipoprotein, plasma carbonyl proteins8-hydroxydeoxyguanosine: decrease
GPx: increase during HBOT
MDA: increase during HBOT
Plasma carbonyl proteins: decrease during HBOT
Oxidized low-density lipoprotein: no significant differences were found
Morabito et al. [43]2011In vivo6 healthy male, well-trained recreational divers11.6 (dive)/2.2 (dive)Immediately post-diveCatalase, hydrogen peroxide, intracellular calcium concentrationCatalase: increase in 2.2 ATA group
Hydrogen peroxide: decrease in 2.2 ATA group
Intracellular calcium concentration: decrease
No significant differences in catalase and hydrogen peroxide levels were found in the 1.6 ATA group
Muth et al. [76]2004In vivo17 healthy male volunteers12.5Immediately post-HBOTCatalase, glutathione, glutathione disulfide, GPx, isoprostanes, MDA, SOD, tail momentIsoprostanes: increase
Catalase, glutathione, glutathione disulfide, GPx, MDA, SOD, tail moment: no significant differences were found
Niu et al. [71]2013In vitroDisc tissue from degenerated lumbar intervertebral discs32.524 h after each sessionNitric oxygenDecrease post-session #2 and post-session #3
Niu et al. [52]2019In vitroDisc tissues from degenerated lumbar intervertebral discs32.512 h post-HBOTInducible NOSDecrease
Niu et al. [72]2011In vitroDisc tissue from degenerated lumbar intervertebral discs32.524 h after each sessionNitric oxygenDecrease post-session #2 and post-session #3
Paprocki et al. [89]2020In vivo23 patients; difficult-to heal skin wounds following mechanical injuries252.5Post-session #1 and #25Catalase, GPx, SOD, TBARSNo significant differences were found
Paprocki et al. [85]2019In vivo40 patients; sudden sensorineural hearing loss142.55 min post-session #1 and post-session #14Catalase, GPx, SOD, TBARSCatalase: decrease post-session #1
GPx: increase post-session #14
SOD: decrease post-session #14
TBARS: increase in the erythrocytes post-session #14
No significant differences in TBARS levels in the plasma were found
Puthucheary et al. [69]2006In vivo15 patients12.4Immediately post-HBOT(exhaled) Nitric oxygenDecrease
Resanovic et al. [53]2019In vivo19 patients; type 1 diabetes mellitus102.4?Free fatty acid, inducible NOS, nitrate/nitriteFree fatty acid: decrease
Inducible NOS: decrease
Nitrate/nitrite: decrease
Rocco et al. [87]2001In vivo15 healthy volunteers12/2.8During HBOT and 30 min post-HBOTTBARSIncreaseOnly pressurization (without breathing 100% O2) has approximately the same effect
Rockswold et al. [79]2010In vivo69 patients; severe traumatic brain injury11.5?IsoprostanesNo significant differences were found
Rossignol et al. [101]2007In vivo18 patients; children with autism401.3/1.5Within 24 h post-HBOTGlutathione disulfideSSGNo significant differences were found
Rothfuss et al. [95]2001In vitroHuman lymphocytes131 h, 4 h, 8 h, 12 h, and 24 h post-HBOTHeme oxygenase-1, tail momentHeme oxygenase-1: increase as of 4 h post-HBOT
Tail moment: increase
Rothfuss et al. [96]2002In vitroHuman lymphocytes11.51 h, 4 h, 8 h, 12 h, and 24 h post-HBOTHeme oxygenase-1, tail momentHeme oxygenase-1: increase as of 4 h post-HBOT
Tail moment: increase
Shaw et al. [66]2009In vitroHuman platelets12.2?Nitrate, nitriteNo significant differences were foundThe level of significance was not determined
Shyu et al. [40]2008In vitroBone marrow-derived human mesenchymal stem cells12.5?ROSIncrease
Sinan et al. [165]2016In vivo33 patients; various disorders202.4Post-session #1 and #20SODNo significant differences were found
Speit et al. [97]2000In vivo14 healthy volunteers12.5Immediately or 1 day post-HBOTcatalase, heme oxygenase-1, SOD, tail momentHeme oxygenase-1: increase
Catalase, SOD, tail moment: no significant differences were found
Sureda et al. [49]2014In vivo9 mail professional divers16 (dive)30 min and 3 h post-diveInducible NOS, MDA, nitrite, nitrotyrosine, nitric oxideNitrite: increase 3 h post-dive
Nitrotyrosine: increase
Nitric oxide: increase
Inducible NOS, MDA: no significant differences were found
Sureda et al. [62]2016In vivo14 patients; chronic non-healing wound202.2Pre- and 2 h post-session #1, #5, and #20Catalase, glutathione reductase, GPx, MDA, myeloperoxidase, nitrite, SODCatalase: increase post-session #1 and post-session #5 in plasma
MDA: decrease pre-session #20 and post-session #20
Myeloperoxidase: decrease post-session #1, post-session #5, and post-session #20
Glutathione reductase, GPx, nitrite, SOD: no significant differences were found
No significant differences were found in catalase levels in erythrocytes
Sureda et al. [55]2009In vivo7 male preprofessional divers15 (dive)Immediately and 3 h post-diveCatalase, glutathione reductase, GPx, MDA, nitrite, protein carbonyl derivates, SODCatalase: increase immediately post-dive in plasma
Nitrite: increase 3 h post-dive
SOD: increase 3 h post-dive in plasma
Glutathione reductase, GPx, MDA, protein carbonyl derivates: no significant differences were found
No significant differences were found in catalase and SOD levels in erythrocytes
Taraldsoy et al. [70]2007In vivo8 patients; chronic radiation-induced injury202.3Post-session #1 and #19(exhaled) Nitric oxideDecrease
Taylor et al. [90]2012In vivo6 healthy, recreationally active, non-smoking male volunteers12.8Within 1 h post-HBOT and 5 h post-HBOTTBARSNo significant differences were found
Teksam et al. [83]2019In vivo54 patients; children with CO poisoning15Within 1 h post-HBOT8-hydroxy-deoxyguanosine, catalase, glutathione, GPx, MDA, plasma carbonyl proteins, SODNo significant differences were found
Tepic et al. [33]2018In vivo50 patients; type 2 diabetes mellitus101.7Post-session #3, #5, #7, and #10Catalase, hydrogen peroxide, nitrite, SOD, superoxide-ion, TBARSCatalase: increase post-session #3 in group without vascular complications and post-session #10 in group with vascular complications
Hydrogen peroxide: decrease post-session #3 in group without vascular complications
Nitrite: increase post-session #3 in group with vascular complications
Superoxide-ion: increase post-session #3 and post-session #10 in group with vascular complications
SOD, TBARS: no significant differences were found
Thom et al. [47]2011In vivo8 patients; diabetes mellitus202?NOSIncrease
Tillmans et al. [98]2019In vitroPeripheral blood mononuclear cells from 49 healthy male subjects14Immediately post-HBOT and 18 h post-HBOTTail momentNo significant differences were found
Uusijärvi et al. [61]2015In vivo19 healthy volunteers12.5During HBOT, 5 min post-HBOT and 30 min post-HBOTNitrate, nitrite, nitric oxygenNitrite: decrease during HBOT and 5 min post-HBOT
NO: decrease in exhaled values
Nitrate: no significant differences were found
No significant differences were found in the NO values in the plasma
Wang et al. [50]2011In vivo77 patients; diabetic foot ulcers202.5?Endothelial NOSNo significant differences were found
Wang et al. [51]2009In vivo74 patients; diabetic foot ulcers302.5?Endothelial NOSNo significant differences were found
Wang et al. [73]2011In vitroDisc tissue from lumbar intervertebral discs32.524 h post-HBOTNitric oxygenDecrease
Wang et al. [48]2020In vivo78 patients; spinal cord injury302?Endothelial NOSIncrease
Yuan et al. [54]2014In vitroAtricular cartilage specimens12.524 h post-HBOTInducible NOSDecrease
Zhou et al. [41]2018In vitroHuman umbilical vein endothelial cells12.8?ROSIncrease
a All studies used a dry exposure in a hyperbaric chamber, unless ‘dive’ is specified. b In minutes (min), hours (h), days, weeks, or months post-HBOT. The baseline measurement point has not been included. ? No information on the moment of sample taking (or just ‘post-HBOT’) was noted in the study.
Table A4. Retrieved results on inflammation markers in detail.
Table A4. Retrieved results on inflammation markers in detail.
StudyYearDesignSubjectsMethodsType Inflammation MarkerOutcomeRemarks
Amount of
Sessions
Maximum Pressure (ATA) aMoment of Sample Taking b
Akcali et al. [104]2018In vivo40 patients; CO poisoning12.46 h post-HBOThs-CRP, IL-6, IL-10No significant differences were found
Alex et al. [127]2005In vivo64 patients; on-pump coronary artery bypass grafting3 (within 24 h)1.5/2.4Preoperative (4 h post-HBOT), 2 h postoperative, and 24 h postoperativeIL-6, IL-8, TNF-αIL-8: decrease preoperative
TNF-α: increase 2 h postoperative
IL-6: no significant differences were found
On-pump coronary artery bypass grafting in the follow-up period
Anguiano-Hernandez et al. [125]2019In vivo18 patients; diabetic foot ulcers201.4Post-session #20IFN-γ, IL-4, IL-6, IL-10, NF-κBIL-6: increase
NF-κB: decrease in the nucleus
IFN-γ, IL-4, IL-10: no significant differences were found
No significant differences in NF-κB levels were seen in the cytosol. The levels of IL-4 were below detection limits.
Aydin et al. [113]2013In vivo48 patients; diabetic foot ulcers302.4?Insulin-like growth factor-1IncreaseThe level of significance was not determined
Baiula et al. [120]2020In vivo30 patients; chronic non-healing wound152.4Immediately post-session #4, #8, #12, and #15 and 1 month post-HBOTIL-1β, TNF-αIL-1β: decrease as of post-session #12
TNF-α: decrease as of post-session #12
Benson et al. [121]2003In vitroPeripheral blood mononuclear cells12.4?IL-1β, TNF-αIL-1β: decrease
TNF-α: decrease
LPS-, lipid A- and PHA-induced IL-1β and TNF-α production was measured
Bent et al. [112]2012In vivo10 children; autism spectrum disorder801.5Post-session #40 and #80Granulocyte-colony stimulating factor, IFN-α, IFN-γ, IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12(p40), IL-12(p70), IL-13, IL-15, IL-17, TNF-αNo significant differences were found
Bosco et al. [4]2014In vivo21 patients; pancreactico-duodenectomy12.5Post-HBOT, 1 day postoperative, and 7 days postoperativeIL-1, IL-6, IL-8, IL-10, IL-12p70, TNF-αIL-6: decrease
IL-10: decrease
IL-1, IL-8, IL-12p70, TNF-α: no significant differences were found
Pancreaticoduodenectomy in the follow-up period
Bosco et al. [35]2018In vivo23 patients; unilateral femoral head necrosis602.5Post-session #15, #30, and #60 and pre-session #31IL-1β, IL-6, TNF-αIL-6: decrease
TNF-α: decrease
IL-1β: no significant differences were found
Chang et al. [102]2020In vivo10 healthy male volunteers12.830 min, 2 days, and 1 week post-HBOTAlbuminNo significant differences were found
Chen et al. [106]2017In vivo38 patients; diabetic foot ulcers202.5Post-session #10, post-session #20, and 2 weeks post-HBOTCRP, erythrocyte sedimentation rateCRP: decrease 2 weeks post-session
Erythrocyte sedimentation rate: decrease 2 weeks post-session
Chen et al. [88]2018In vivo50 patients; acute non-cardioembolic stroke12.51 month post-HBOThs-CRPNo significant differences were found
Chen et al. [67]2007In vivo61 patients; diabetes mellitus type 232.5Immediately post-session #1 and #3Insulin-like growth factor-1, IL-8No significant differences were found
Chong et al. [118]2013In vivo17 patients; thermal burns22.4?IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12(p70), IL-13, TNF-αIFN-γ: increase
IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12(p70), IL-13: no significant differences were found
Outcome on TNF-α is not reported
Dhamodharan et al. [45]2019In vivo37 patients; diabetic foot ulcer252.220 days after the first HBOT-sessionCRP, IFN-γ, IL-1α, IL-8, IL-10, TNF-αCRP: decrease
IFN-γ: decrease
IL-1α: increase
IL-8: increase
IL-10: increase
TNF-α: no significant differences were found
Fan et al. [122]2020In vivo122 patients; Parkinson’s disease dementia4020 MPA?IL-1β, IL-6IL-1β: decrease
IL-6: decrease
Fildissis et al. [128]2004In vitroBlood samples from 16 healthy volunteers12.4?IL-6, IL-8, TNF-αNo significant differences were found
Guggino et al. [133]2019In vivo36 patients; primary fibromyalgia4021 month post-HBOTIFN-γ, IL-9, IL-17, IL-22, TNF-αIFN-γ: decrease
TNF-α: decrease
IL-9, IL-17, IL-22: no significant differences were found
Hao et al. [111]2020In vivo30 patients; plastic surgery7224 h post-HBOTGranulocyte-colony stimulating factor, IFN-α2, IFN-γ, IL-1α, IL-1β, IL-1Ra, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12p40, IL-12p70, IL-13, IL-15, IL-17, TNF-αIL-1Ra: increase
IL-12p40: decrease
Granulocyte-colony stimulating factor, IFN-α2, IFN-γ, IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12p70, IL-13, IL-15, IL-17, TNF-α: no significant differences were found
The samples were taken during surgery
Hedetoft et al. [114]2020In vivo39 patients; diabetes302.4Post-session #30 and after 90 days follow-upInsuline-like growth factor-1Decrease after 90 days follow-up in the diabetic groupNo significant differences in IGF-1 values were seen in the non-diabetic group
Hou et al. [129]2019In vivo132 patients; brain tumor101.8?IL-6, TNF-αIL-6: decrease
TNF-α: decrease
Irawan et al. [116]2018In vivo36 patients; diabetic foot ulcers202.4Post-session #20AlbuminIncrease
Irawan et al. [117]2018In vivo30 patients; diabetic foot ulcers102.4?AlbuminNo differences were foundThe level of significance was not determined
Karadurmus et al. [103]2010In vivo28 patients; diabetic foot ulcers302.4Post-session #10, #20, and #30hs-CRPDecrease
Kendall et al. [46]2012In vitroHuman umbilical vein endothelial cells12.45 h and 22.5 h post-HBOTIL-8Decrease 5 h post-HBOT
Li et al. [84]2017In vivo78 patients; chronic diabetic woundsBy average 482.430 days after the first sessionCRP, TNF-αCRP: decrease
TNF-α: decrease
Li et al. [58]2018In vivo115 patients; coronary artery disease with drug-eluting stents242?hs-CRPDecrease
Li et al. [59]2019In vivo115 patients; coronary artery disease with coronary stent implantation242?hs-CRPDecrease
Li et al. [60]2018In vivo98 patients; slow coronary flow242?hs-CRPDecrease
Lin et al. [130]2018In vivo57 patients; peripheral arterial occlusive disease10–152.5Post-session #3 and #5IL-6Decrease
Liu et al. [137]2020In vivo140 patients; unilateral idiopathic sudden sensorineural hearing loss152?NF-κB, TNF-αNF-κB: decrease
TNF-α: decrease
MacKenzie et al. [126]2000In vitroPeripheral blood mononuclear cells1 (duration: 48 h)1.77 days post-HBOTIFN-γ, IL-4, IL-10No significant differences were foundThe IL-4 values were below detection limits
Madden et al. [139]2011In vivo10 healthy male volunteers12.8Immediately post-HBOT and 4 h post-HBOTNF-κBIncrease 4 h post-HBOT
Nasole et al. [135]2014In vivo27 patients; chronic leg wounds402.57 and 14 days after the first HBOT-sessionTNF-αNo significant differences were found
Niu et al. [71]2013In vitroDisc tissue from degenerated lumbar intervertebral discs32.524 h after each sessionIL-1βDecrease
Niu et al. [52]2019In vitroAbnormal disc tissues from degenerated lumbar intervertebral discs32.51 h post-session #3NF-κBDecrease in the nucleus
Niu et al. [72]2001In vitroDisc tissue from degenerated lumbar intervertebral discs32.524 h after each sessionIL-1βDecrease
Resanovic et al. [53]2019In vivo19 patients; type 1 diabetes mellitus102.4?CRP, NF-κBCRP: decrease
NF-κB: decrease
Rocco et al. [87]2001In vivo15 healthy volunteers12.8During HBOT and 30 min post-HBOTIL-1β, IL-6, TNF-αTNF-α: increase during HBOT
IL-1β, IL-6: no significant differences were found
Romero-valdovinos et al. [115]2011In vitroDermal fibroblasts303?Insulin-like growth factor-1Decrease in the keloid fibroblast groupThe nonkeloid fibroblasts did not express IGF-1
Rosario et al. [131]2018In vivo6 patients; ischemic stroke402?IL-6, TNF-αIL-6: decrease
TNF-α: decrease
Rossignol et al. [101]2007In vivo18 patients; children with autism401.3/1.5Within 24 h post-HBOTCRPDecreaseIn the group with lower CRP baseline levels no decrease in CRP concentrations was seen
Rothfuss et al. [96]2002In vitroHuman lymphocytes21.5/34 h, 8 h, and 24 h post-HBOTFerritinNo significant differences were found
Schnittger et al. [110]2004In vivo8 patients; CO poisoning3 (within 24 h)2.8Before and after each sessionGranulocyte-colony stimulating factor, IL-8Granulocyte-colony stimulating factor: increase pre- and post-session #2 in the patient group
IL-8: no significant differences were found
No significant differences in granulocyte-colony stimulating factor values were seen in the control group
Semadi et al. [138]2019In vivo32 patients; diabetic foot ulcer202.4Post-session #20TNF-αDecrease
Song et al. [132]2018In vivo134 patients; keloid surgery and radiotherapy142Post-operation #2IL-6, NF-κB, TNF-αIL-6: decrease
NF-κB: decrease
TNF-α: decrease
Keloid surgery and radiotherapy in the follow-up period
Sun et al. [119]2018In vivo80 patients; brain injury2028 h, 24 h, 48 h, and 72 h post-HBOTIL-1β, TNF-αTNF-α: decrease
IL-1β: no significant differences were found
Baseline values of TNF-α were significantly higher in the HBOT-group compared to the control group
Sun et al. [140]2019In vivo79 patients; acute spinal cord injury302Post session #1, #3, #7, #10, and #30NF-κBDecrease post-session #3, #7, #10, and #30
Sureda et al. [62]2016In vivo14 patients; chronic non-healing wound202.2Pre- and 2 h post-session #1, #5, and #20IL-6Increase 2 h post-session #1, #5, and #20
Top et al. [107]2007In vivo38 patients; type 2 diabetes mellitus?22 weeks after the first HBOT-sessionCRP, erythrocyte sedimentation rateCRP: decrease
Erythrocyte sedimentation rate: increase
Vezzani et al. [105]2016In vivo30 patients; CO poisoning12.5/2.8Immediately post-HBOTCRP, IL-6, IL-8, IL-10, TNF-αIL-8: decrease in the control group
IL-10: decrease in the patient group
TNF-α: decrease
CRP, IL-6: no significant differences were found
No significant differences regarding IL-8 and IL-10 were seen in respectively the patient group and the control group
Wang et al. [134]2011In vitroHuman coronary artery endothelial cells12.5?TNF-αIncrease during HBOTThe TNF-α values returned to the baseline values before the HBOT ended
Wang et al. [73]2011In vitroDisc tissue from lumbar intervertebral discs12.548 h, 96 h, and 144 h post-HBOTIL-1βDecrease
Weisz et al. [124]1997In vivo7 patients; perianal Crohn’s disease20/402.5Immediately post-session #1, 24 h post-session #1, 20 h post-session #2, and 20 h post-session #20IL-1, IL-6, TNF-αIL-1: decrease immediately and 24 h post-session #1 and 20 h post-session #2
IL-6: decrease immediately and 24 h post-session #1 and 20 h post-session #2
TNF-α: decrease immediately post-session #1 and 20 h post-session #2
Wilkinson et al. [123]2015In vivo19 male volunteers; overweight/obese42During HBOT, immediately post-session #4, and 24 h post-session #4IL-1Ra, IL-6, IL-18, TNF-αIL-6: increase during HBOT and immediately post-session #4 in the non-diabetes group
TNF-α: decrease 24 h post-session #4
IL-1Ra, IL-18: no significant differences were found
Xie et al. [109]2007In vivo60 patients; craniocerebral injury102.5Within 24 h post-HBOTCRPDecrease
Yildiz et al. [108]2016In vivo43 patients; hidradenitis suppurativa202.4Post-session #20 and 6 weeks post-HBOTCRP, erythrocyte sedimentation rateCRP: decrease
Erythrocyte sedimentation rate: decrease
Yoshinoya et al. [136]2020In vitroAdipose-derived stem cells52/390 min before and immediately after each sessionTNF-αNo significant differences were foundThe TNF-α values were below detection limits
a All studies used a hyperbaric chamber for pressurization. b In minutes (min), hours (h), days, weeks, or months post-HBOT. The baseline measurement point has not been included. ? No information on the moment of sample taking (or just ‘post-HBOT’) was noted in the study.
Table A5. Retrieved results on angiogenesis markers in detail.
Table A5. Retrieved results on angiogenesis markers in detail.
StudyYearDesignSubjectsMethodsType Angiogenesis MarkerOutcomesRemarks
Amount of
Sessions
Maximum Pressure (ATA) aMoment of Sample Taking b
Anguiano-Hernandez et al. [125]2019In vivo18 patients; diabetic foot ulcers201.4Post-session #20HIF-1α, insulin-like growth factor binding protein-3, VEGFInsulin-like growth factor binding protein-3: increase in nucleus and fibroblast
VEGF: increase in the cytosol
HIF-1α: no significant differences were found
No significant differences in insulin-like growth factor binding protein-3 and VEGF levels were found in the cytoplasm and nucleus, respectively
Bent et al. [112]2012In vivo10 children; autism spectrum disorder801.5Post-session #40 and #80Granulocyte-macrophage colony-stimulating factor, TGF-β1, TGF-β2No significant differences were found
Chang et al. [102]2020In vivo10 healthy male volunteers12.830 min, 2 days, and 1 week post-HBOTFGF21No significant differences were found
Chen et al. [67]2007In vivo61 patients; diabetes mellitus type 232.5Immediately post-session #1 and #3Insulin-like growth factor-2, insulin-like growth factor binding protein-1, insulin-like growth factor binding protein-3Insulin-like growth factor binding protein-1: decrease post-session #1 and (less prominent) post-session #3
Insulin-like growth factor-2, insulin-like growth factor binding protein-3: no differences were found
No significance levels were determined
Cheung et al. [37]2018In vitroUmbilical cord blood enriched with CD34-cells12.524 h post-HBOTErythropoietin-receptorNo significant differences were found
Chong et al. [118]2013In vivo17 patients; thermal burns22.4?VEGFNo significant differences were found
Dhamodharan et al. [45]2019In vivo37 patients; diabetic foot ulcer252.220 days after the first HBOT-sessionEGF, FGF-2, platelet-derived growth factor, VEGFEGF: increase
FGF-2: increase
VEGF: increase
Platelet-derived growth factor: no significant differences were found
Grimberg-Peters et al. [44]2016In vitroNeutrophils from severely injured patients and healthy volunteers12?ERK, p38 MAPKp38 MAPK: decrease
ERK: no significant differences were found
The decrease in p38 MAPK levels was only found after 3h of stimulation with PMA
Hao et al. [111]2020In vivo30 patients; plastic surgery7224 h post-HBOTEGF, FGF-2, granulocyte-macrophage colony-stimulating factor, platelet-derived growth factor-AA, platelet-derived growth factor-BB, TGF-α, VEGFPlatelet-derived growth factor-BB: decrease
EGF, FGF-2, granulocyte-macrophage colony-stimulating factor, platelet-derived growth factor-AA, TGF-α, VEGF: no significant differences were found
The samples were taken during surgery.
Jung et al. [143]2010In vivo86 patients; acute hearing loss/tinnitus101.551, 2, 5, and 10 days after the first HBOT-sessionbFGF, VEGFbFGF: decrease
VEGF: no significant differences were found
Kang et al. [147]2004In vitroFibroblast primary cell lines71.5/2/2.5/31 day, 3 days, 5 days, and 7 days after the first HBOT-sessionbFGF, TGF-β1, VEGFNo significant differences were foundAdministration of 100% oxygen significantly increased the bFGF levels at day 1, yet pressurization had no extra effect. The TGF-β1 values were below detection limits.
Kunnavatana et al. [148]2005In vitroFibroblast cell line721 day, 3 days, 5 days, and 7 days after the first HBOT-sessionbFGF, TGF-β1, VEGFNo significant differences were found
Lee et al. [142]2006In vitroHuman umbilical vein endothelial cells12.5 ERK, VEGFERK: increase
VEGF: increase
Li et al. [84]2017In vivo78 patients; chronic diabetic woundsBy average 482.430 days after the first HBOT-sessionAng-2, VEGFAng-2: increase
VEGF: increase
Lin et al. [130]2018In vivo57 patients; peripheral arterial occlusive disease10 of 152.5Post-session #3 and #5Hematopoietic growth factor, HIF-1α, stromal cell-derived factor-1α, VEGFHematopoietic growth factor: increase
HIF-1α: decrease
Stromal cell-derived factor-1α: increase
VEGF: increase
Lin et al. [144]2002In vitroHuman umbilical vein endothelial cells12.5?Ang-1, Ang-2, Tie-2, VEGFNo significant differences were foundAdministration of 100% oxygen significantly increased the Ang-2 levels, yet pressurization had no extra effect
Mutzbauer et al. [145]2015In vivo16 divers31.4 (dive)Within 1 h pre- and post-diveErythropoietinDecrease post-dive #2 and #3
Nasole et al. [135]2014In vivo27 patients; chronic leg wounds402.57 and 14 days after the first HBOT-sessionEGF, VEGFNo significant differences were found
Niu et al. [71]2013In vitroDisc tissue from degenerated lumbar intervertebral discs32.530 min and 60 min post-session #3ERK1/2, p38 MAPKERK1/2: decrease
p38 MAPK: decrease
Phosphorylation of p38 MAPK and ERK has been measured
Niu et al. [52]2019In vitroAbnormal disc tissues from degenerated lumbar intervertebral discs32.5ERK1/2, p38 MAPK: 15 min and 30 min post-session #3
MMP-3, MPP-9, MMP-13: 12 h after each session
ERK1/2, MMP-3, MMP-9, MMP-13, p38 MAPKERK1/2: decrease 30 min post-session #3
MMP-3: decrease post-session #2 and #3
MMP-9: decrease post-session #2 and #3
MMP-13: decrease post-session #2 and #3
p38 MAPK: decrease
Phosphorylation of p38 MAPK and ERK has been measured
Niu et al. [72]2011In vitroDisc tissue from degenerated lumbar intervertebral discs32.5MMP-3: 24 h after each session
p38 MAPK: 15 min, 30 min, and 60 min post-session #3
MMP-3, p38 MAPKMMP-3: decrease post-session #3
p38 MAPK: decrease
Phosphorylation of p38 MAPK and ERK has been measured
Resanovic et al. [53]2019In vivo19 patients; type 1 diabetes mellitus102.4?Akt, ERK1/2Akt: decrease
ERK1/2: decrease
Romero-valdovinos et al. [115]2011In vitroDermal fibroblasts303?TGF-βTGF-β: decrease
Semadi et al. [138]2019In vivo32 patients; diabetic foot ulcer202.4Post-session #20VEGFIncrease
Shyu et al. [40]2008In vitroBone marrow-derived human mesenchymal stem cells12.51 h, 2 h, 4 h, and 6 h post-HBOTPGFIncreaseThe increase in PGF levels was higher at 1h and 2h post-HBOT compared to 4h and 6h post-HBOT
Song et al. [132]2018In vivo134 patients; keloid surgery and radiotherapy142Post-operation #2HIF-1α, VEGFHIF-1α: decrease
VEGF: decrease
Keloid surgery and radiotherapy in the follow-up period
Sureda et al. [62]2016In vivo14 patients; chronic non-healing wound202.2Pre- and 2 h post-session #1, #5, and #20VEGFIncrease post-session #1, post-session #5, and post-session #20
Thom et al. [47]2011In vivo8 patients; diabetes mellitus202Pre- and post-session #1, #10, and #20HIF-1αDecrease post-session #1, #10, and #20No significant differences in HIF-1α levels were found pre-session
Tra et al. [141]2014In vitroTissue-engineered mucosa and human umbilical vein endothelial cells1/3/52.4Immediately post-HBOTbFGF, hematopoietic growth factor, keratinocyte growth factor, PGF, VEGFbFGF: an increase in the one-session group and a decrease in the three- and five-session group
Hematopoietic growth factor: increase in the one-session group
Keratinocyte growth factor: increase in the one- and five-session group
PGF: an increase in the one- and five-session group and a decrease in the three-session group
VEGF: increase in the five-session group
Wang et al. [50]2011In vivo77 patients; diabetic foot ulcers202.5?EGF, VEGFNo significant differences were found
Wang et al. [51]2009In vivo74 patients; diabetic foot ulcers302.5?VEGFNo significant differences were found
Wang et al. [48]2020In vivo78 patients; spinal cord injury302?Akt, PI3KAkt: increase
PI3K: increase
Yoshinoya et al. [136]2020In vitroAdipose-derived stem cells52/390 min before and immediately after each sessionEGF, hematopoietic growth factor, TGF-βTGF-β: decrease post-session #3 in the 2 ATA group and post-session #4 in the 3 ATA group
EGF, hematopoietic growth factor: no significant differences were found
The EGF values were below detection limits
Yuan et al. [54]2014In vitroAtricular cartilage specimens12.524 h post-HBOTMMP-3Decrease
a All studies used a dry exposure in a hyperbaric chamber, unless ‘dive’ is specified. b In minutes (min), hours (h), days, or months post-HBOT. The baseline measurement point has not been included. ? No information on the moment of sample taking (or just ‘post-HBOT’) was noted in the study.

References

  1. Society, U.H.M. Hyperbaric Oxygen Therapy Indications, 14th ed.; Moon, R., Ed.; Best Publishing Company: North Palm Beach, FL, USA, 2019. [Google Scholar]
  2. Tibbles, P.M.; Edelsberg, J.S. Hyperbaric-oxygen therapy. N. Engl. J. Med. 1996, 334, 1642–1648. [Google Scholar] [CrossRef]
  3. Society, U.A.H.M. UHMS Guidelines for credentialing, privileging and supervision of hyperbaric oxygen therapy in the U.S.A. Undersea Hyperb. Med. 2018, 45, 117–127. [Google Scholar] [CrossRef]
  4. Bosco, G.; Casarotto, A.; Nasole, E.; Camporesi, E.; Salvia, R.; Giovinazzo, F.; Zanini, S.; Malleo, G.; Di Tano, A.; Rubini, A.; et al. Preconditioning with hyperbaric oxygen in pancreaticoduodenectomy: A randomized double-blind pilot study. Anticancer Res. 2014, 34, 2899–2906. [Google Scholar]
  5. Friedman, T.; Menashe, S.; Landau, G.; Sherf, M.; Wiser, I.; Seligman, Y.; Friedman, M.; Hadanny, A.; Efrati, S.; Heller, L. Hyperbaric oxygen preconditioning can reduce postabdominoplasty complications: A retrospective cohort study. Plast. Reconstr. Surg. Glob. Open 2019, 7, e2417. [Google Scholar] [CrossRef]
  6. Li, Y.; Dong, H.; Chen, M.; Liu, J.; Yang, L.; Chen, S.; Xiong, L. Preconditioning with repeated hyperbaric oxygen induces myocardial and cerebral protection in patients undergoing coronary artery bypass graft surgery: A prospective, randomized, controlled clinical trial. J. Cardiothorac. Vasc. Anesth. 2011, 25, 908–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Yogaratnam, J.Z.; Laden, G.; Guvendik, L.; Cowen, M.; Cale, A.; Griffin, S. Hyperbaric oxygen preconditioning improves myocardial function, reduces length of intensive care stay, and limits complications post coronary artery bypass graft surgery. Cardiovasc. Revascularization Med. 2010, 11, 8–19. [Google Scholar] [CrossRef]
  8. Toner, A.; Hamilton, M. The long-term effects of postoperative complications. Curr. Opin. Crit. Care 2013, 19, 364–368. [Google Scholar] [CrossRef] [PubMed]
  9. Pinto, A.; Faiz, O.; Davis, R.; Almoudaris, A.; Vincent, C. Surgical complications and their impact on patients’ psychosocial well-being: A systematic review and meta-analysis. BMJ Open 2016, 6, e007224. [Google Scholar] [CrossRef] [Green Version]
  10. Carey, K.; Stefos, T.; Zhao, S.; Borzecki, A.M.; Rosen, A.K. Excess costs attributable to postoperative complications. Med. Care Res. Rev. 2011, 68, 490–503. [Google Scholar] [CrossRef]
  11. Camporesi, E.M.; Bosco, G. Mechanisms of action of hyperbaric oxygen therapy. Undersea Hyperb. Med. 2014, 41, 247–252. [Google Scholar]
  12. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [Green Version]
  13. Fosen, K.M.; Thom, S.R. Hyperbaric oxygen, vasculogenic stem cells, and wound healing. Antioxid. Redox Signal. 2014, 21, 1634–1647. [Google Scholar] [CrossRef] [Green Version]
  14. Eken, A.; Aydin, A.; Sayal, A.; Ustundag, A.; Duydu, Y.; Dundar, K.; Aydın, A. The effects of hyperbaric oxygen treatment on oxidative stress and SCE frequencies in humans. Clin. Biochem. 2005, 38, 1133–1137. [Google Scholar] [CrossRef]
  15. Speit, G.; Dennog, C.; Radermacher, P.; Rothfuss, A. Genotoxicity of hyperbaric oxygen. Mutat. Res. Mol. Mech. Mutagen. 2002, 512, 111–119. [Google Scholar] [CrossRef]
  16. Chelombitko, M.A. Role of reactive oxygen species in inflammation: A minireview. Mosc. Univ. Biol. Sci. Bull. 2018, 73, 199–202. [Google Scholar] [CrossRef] [Green Version]
  17. Hehenberger, K.; Brismar, K.; Lind, F.; Kratz, G. Dose-dependent hyperbaric oxygen stimulation of human fibroblast proliferation. Wound Repair Regen. 1997, 5, 147–150. [Google Scholar] [CrossRef]
  18. Chiang, I.-H.; Chen, S.-G.; Huang, K.-L.; Chou, Y.-C.; Dai, N.-T.; Peng, C.-K. Adjunctive hyperbaric oxygen therapy in severe burns: Experience in Taiwan formosa water park dust explosion disaster. Burns 2017, 43, 852–857. [Google Scholar] [CrossRef] [PubMed]
  19. Dimitrijevich, S.D.; Paranjape, S.; Wilson, J.R.; Gracy, R.W.; Mills, J.G. Effect of hyperbaric oxygen on human skin cells in culture and in human dermal and skin equivalents. Wound Repair Regen. 1999, 7, 53–64. [Google Scholar] [CrossRef] [PubMed]
  20. Granowitz, E.V.; Tonomura, N.; Benson, R.M.; Katz, D.M.; Band, V.; Makari-Judson, G.P.; Osborne, B.A. Hyperbaric oxygen inhibits benign and malignant human mammary epithelial cell proliferation. Anticancer Res. 2005, 25, 3833–3842. [Google Scholar] [PubMed]
  21. Hibbs, H.W.; Harasym, M.P.; Bansal, D.; Stewart, J. Effects of a single hyperbaric oxygen exposure on haematocrit, prothrombin time, serum calcium, and platelet count. Diving Hyperb. Med. South Pac. Underw. Med. Soc. 2007, 37, 143–145. [Google Scholar]
  22. Hollander, D.A.; Hakimi, M.Y.; Hartmann, A.; Wilhelm, K.; Windolf, J. The influence of hyperbaric oxygenation (HBO) on proliferation and differentiation of human keratinocyte cultures in vitro. Cell Tissue Bank. 2000, 1, 261–269. [Google Scholar] [CrossRef]
  23. Jüttner, B.; Scheinichen, D.; Bartsch, S.; Heine, J.; Ruschulte, H.; Elsner, H.A.; Franko, W.; Jaeger, K. Lack of toxic side effects in neutrophils following hyperbaric oxygen. Undersea Hyperb. Med. 2003, 30, 305–311. [Google Scholar] [PubMed]
  24. Kairuz, E.; Upton, Z.; Dawson, R.A.; Malda, J. Hyperbaric oxygen stimulates epidermal reconstruction in human skin equivalents. Wound Repair Regen. 2007, 15, 266–274. [Google Scholar] [CrossRef] [Green Version]
  25. Li, H.; Zhao, D.; Diao, M.; Yang, C.; Zhang, Y.; Lv, Y.; Zhao, J.; Pan, S. Hyperbaric oxygen treatments attenuate the Neutrophil-to-Lymphocyte ratio in patients with idiopathic sudden sensorineural hearing loss. Otolaryngol. Neck Surg. 2015, 153, 606–612. [Google Scholar] [CrossRef] [PubMed]
  26. Melcher, C.; Sievers, B.; Höchsmann, N.; Düren, F.; Jansson, V.; Müller, P.E. Effect of hyperbaric oxygen on proliferation and gene expression of human chondrocytes: An in vitro study. Cartilage 2019, 10, 459–466. [Google Scholar] [CrossRef] [PubMed]
  27. Monaca, E.; Strelow, H.; Jüttner, T.; Hoffmann, T.; Potempa, T.; Windolf, J.; Winterhalter, M. Assessment of hemostaseologic alterations induced by hyperbaric oxygen therapy using point-of-care analyzers. Undersea Hyperb. Med. 2014, 41, 17–26. [Google Scholar]
  28. Passavanti, G.; Tanasi, P.; Brauzzi, M.; Pagni, M.; Norgini, E.; Aloisi, A. can hyperbaric oxygenation therapy (HOT) modify the blood testosterone concentration? Urol. J. 2010, 77, 52–56. [Google Scholar] [CrossRef]
  29. Ravaioli, M.; Baldassare, M.; Vasuri, F.; Pasquinelli, G.; Laggetta, M.; Valente, S.; De Pace, V.; Neri, F.; Siniscalchi, A.; Zanfi, C.; et al. Strategies to restore adenosine triphosphate (ATP) level after more than 20 hours of cold ischemia time in human marginal kidney grafts. Ann. Transplant. 2018, 23, 34–44. [Google Scholar] [CrossRef] [PubMed]
  30. Roberts, G.P.; Harding, K.G. Stimulation of glycosaminoglycan synthesis in cultured fibroblasts by hyperbaric oxygen. Br. J. Dermatol. 1994, 131, 630–633. [Google Scholar] [CrossRef] [PubMed]
  31. Roberts, G.P.; Qu, B.; Harding, K.G. The effects of hyperbaric oxygen on cultured fibroblasts. J. Wound Care 1994, 3, 189–193. [Google Scholar] [CrossRef]
  32. Vezzani, G.; Iezzi, M.; Rizzato, A.; Quartesan, S.; Mangar, D.; Camporesi, E.M.; Paganini, M.; Bosco, G. Effects of hyperbaric oxygen exposure on mobilization of endothelial progenitor cells in healthy volunteers. Acta Med. Mediterr. 2017, 33, 801–805. [Google Scholar] [CrossRef]
  33. Tepić, S.; Petković, A.; Srejović, I.; Jeremić, N.; Zivković, V.; Loncarević, S.; Bradić, J.; Jakovljević, V.; Zivkovć, M. Impact of hyperbaric oxygenation on oxidative stress in diabetic patients. Undersea Hyperb. Med. Soc. 2018, 45, 9–17. [Google Scholar] [CrossRef]
  34. Ferrer, M.D.; Sureda, A.; Batle, J.M.; Tauler, P.; Tur, J.A.; Pons, A. Scuba diving enhances endogenous antioxidant defenses in lymphocytes and neutrophils. Free. Radic. Res. 2007, 41, 274–281. [Google Scholar] [CrossRef] [PubMed]
  35. Bosco, G.; Vezzani, G.; Mrakic-Sposta, S.; Rizzato, A.; Enten, G.; Abou-Samra, A.; Malacrida, S.; Quartesan, S.; Vezzoli, A.; Camporesi, E. Hyperbaric oxygen therapy ameliorates osteonecrosis in patients by modulating inflammation and oxidative stress. J. Enzym. Inhib. Med. Chem. 2018, 33, 1501–1505. [Google Scholar] [CrossRef] [Green Version]
  36. Chen, C.-H.; Chien, M.-Y.; Liang, Y.-C.; Liu, D.-Z.; Hu, M.-L. An in vitro hyperbaric oxygen system for evaluation of free radical damage and protection by catechins on hemorheological parameters. Clin. Hemorheol. Microcirc. 2011, 48, 211–221. [Google Scholar] [CrossRef] [PubMed]
  37. Cheung, K.Y.; Berry, A.; Li, D.; Aljitawi, O.S. Hyperbaric oxygen treatment effects on in vitro cultured umbilical cord blood CD34 (+) cells. Cytotherapy 2018, 20, 87–94. [Google Scholar] [CrossRef] [PubMed]
  38. Gröger, M.; Öter, S.; Simkova, V.; Bolten, M.; Koch, A.; Warninghoff, V.; Georgieff, M.; Muth, C.-M.; Speit, G.; Radermacher, P. DNA damage after long-term repetitive hyperbaric oxygen exposure. J. Appl. Physiol. 2009, 106, 311–315. [Google Scholar] [CrossRef] [Green Version]
  39. Lin, H.-I.; Chu, S.-J.; Perng, W.-C.; Wu, C.-P.; Lin, Z.-Y.; Huang, K.-L. Hyperbaric oxygen attenuates cell growth in skin fibroblasts cultured in a high-glucose medium. Wound Repair Regen. 2008, 16, 513–519. [Google Scholar] [CrossRef]
  40. Shyu, K.-G.; Hung, H.-F.; Wang, B.-W.; Chang, H. Hyperbaric oxygen induces placental growth factor expression in bone marrow-derived mesenchymal stem cells. Life Sci. 2008, 83, 65–73. [Google Scholar] [CrossRef]
  41. Zhou, Q.; Huang, G.; Yu, X.; Xu, W. A novel approach to estimate ROS origination by hyperbaric oxygen exposure, targeted probes and specific inhibitors. Cell. Physiol. Biochem. 2018, 47, 1800–1808. [Google Scholar] [CrossRef]
  42. Kendall, A.C.; Whatmore, J.L.; Winyard, P.; Smerdon, G.R.; Eggleton, P. Hyperbaric oxygen treatment reduces neutrophil-endothelial adhesion in chronic wound conditions through S-nitrosation. Wound Repair Regen. 2013, 21, 860–868. [Google Scholar] [CrossRef] [PubMed]
  43. Morabito, C.; Bosco, G.; Pilla, R.; Corona, C.; Mancinelli, R.; Yang, Z.; Camporesi, E.M.; Fanò, G.; Mariggiò, M.A. Effect of pre-breathing oxygen at different depth on oxidative status and calcium concentration in lymphocytes of scuba divers. Acta Physiol. 2010, 202, 69–78. [Google Scholar] [CrossRef] [PubMed]
  44. Grimberg-Peters, D.; Büren, C.; Windolf, J.; Wahlers, T.; Paunel-Görgülü, A. Hyperbaric oxygen reduces production of reactive oxygen species in neutrophils from polytraumatized patients yielding in the inhibition of p38 MAP kinase and downstream pathways. PLoS ONE 2016, 11, e0161343. [Google Scholar] [CrossRef]
  45. Dhamodharan, U.; Karan, A.; Sireesh, D.; Vaishnavi, A.; Somasundar, A.; Rajesh, K.; Ramkumar, K.M. Tissue-specific role of Nrf2 in the treatment of diabetic foot ulcers during hyperbaric oxygen therapy. Free. Radic. Biol. Med. 2019, 138, 53–62. [Google Scholar] [CrossRef]
  46. Kendall, A.C.; Whatmore, J.L.; Harries, L.; Winyard, P.G.; Smerdon, G.R.; Eggleton, P. Changes in inflammatory gene expression induced by hyperbaric oxygen treatment in human endothelial cells under chronic wound conditions. Exp. Cell Res. 2012, 318, 207–216. [Google Scholar] [CrossRef] [PubMed]
  47. Thom, S.R.; Milovanova, T.N.; Yang, M.; Bhopale, V.M.; Sorokina, E.M.; Uzun, G.; Malay, D.S.; Dpm, M.A.T.; Hardy, K.R.; Lambert, D.S.; et al. Vasculogenic stem cell mobilization and wound recruitment in diabetic patients: Increased cell number and intracellular regulatory protein content associated with hyperbaric oxygen therapy. Wound Repair Regen. 2011, 19, 149–161. [Google Scholar] [CrossRef] [Green Version]
  48. Wang, L.; Sun, H.; Fu, S.; Shao, S.; Hou, H.; Liu, X. The role of pi3k/akt/enos pathway in spinal cord injury and the recovery of motor function. Int. J. Clin. Exp. Med. 2020, 13, 4960–4965. [Google Scholar]
  49. Sureda, A.; Batle, J.M.; Capó, X.; Martorell, M.; Córdova, A.; Tur, J.A.; Pons, A. Scuba diving induces nitric oxide synthesis and the expression of inflammatory and regulatory genes of the immune response in neutrophils. Physiol. Genom. 2014, 46, 647–654. [Google Scholar] [CrossRef]
  50. Wang, C.-J.; Ko, J.-Y.; Kuo, Y.-R.; Yang, Y.-J. Molecular changes in diabetic foot ulcers. Diabetes Res. Clin. Pract. 2011, 94, 105–110. [Google Scholar] [CrossRef]
  51. Wang, C.-J.; Kuo, Y.-R.; Wu, R.-W.; Liu, R.-T.; Hsu, C.-S.; Wang, F.-S.; Yang, K.D. extracorporeal shockwave treatment for chronic diabetic foot ulcers. J. Surg. Res. 2009, 152, 96–103. [Google Scholar] [CrossRef] [PubMed]
  52. Niu, C.-C.; Lin, S.-S.; Yuan, L.-J.; Lu, M.-L.; Ueng, S.W.N.; Yang, C.-Y.; Tsai, T.-T.; Lai, P.-L. Upregulation of miR-107 expression following hyperbaric oxygen treatment suppresses HMGB1/RAGE signaling in degenerated human nucleus pulposus cells. Arthritis Res. Ther. 2019, 21, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Resanovic, I.; Gluvic, Z.; Zaric, B.; Sudar-Milovanovic, E.; Jovanovic, A.; Milacic, D.; Isakovic, R.; Isenovic, E.R. Early Effects of hyperbaric oxygen on inducible nitric oxide synthase activity/expression in lymphocytes of Type 1 diabetes patients: A prospective pilot study. Int. J. Endocrinol. 2019, 2019, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Yuan, L.-J.; Niu, C.-C.; Lin, S.-S.; Yang, C.-Y.; Chan, Y.-S.; Chen, W.-J.; Ueng, S.W. Effects of low-intensity pulsed ultrasound and hyperbaric oxygen on human osteoarthritic chondrocytes. J. Orthop. Surg. Res. 2014, 9, 5. [Google Scholar] [CrossRef] [Green Version]
  55. Sureda, A.; Ferrer, M.D.; Batle, J.M.; Tauler, P.; Tur, J.A.; Pons, A. Scuba diving increases erythrocyte and plasma antioxidant defenses and spares no without oxidative damage. Med. Sci. Sports Exerc. 2009, 41, 1271–1276. [Google Scholar] [CrossRef] [PubMed]
  56. Kozakiewicz, M.; Kedziora-Kornatowska, K.; Kaczerska, D.; Siermontowski, P.; Olszanski, R.; Krefft, K. Influence of exposure in hyperbaric chambers on selected parametersof oxidative stress in professional divers. Undersea Hyperb. Med. 2018, 45, 49–54. [Google Scholar] [CrossRef]
  57. Boykin, J.V.; Baylis, C. Hyperbaric oxygen therapy mediates increased nitric oxide production associated with wound healing. Adv. Ski. Wound Care 2007, 20, 382–389. [Google Scholar] [CrossRef] [Green Version]
  58. Li, Y.; Hao, Y.; Wang, T.; Wei, L.; Wang, W.; Liang, Y.; Guo, X. The effect of hyperbaric oxygen therapy on myocardial perfusion after the implantation of drug-eluting stents. Ann. Clin. Lab. Sci. 2018, 48, 158–163. [Google Scholar]
  59. Li, Y.; Hao, Y.-F.; Wang, T.; Zhang, J.-F.; Liang, Y.; Xiao, W.-L.; Guo, X. Hyperbaric oxygen may improve vascular endothelial function in patients undergoing coronary stent implantation. Undersea Hyperb. Med. 2019, 46, 145–152. [Google Scholar] [CrossRef]
  60. Li, Y.; Zhang, H.; Liang, Y.; Wang, W.; Xu, T.; Zhang, J.; Xiao, W.; Wang, T. Effects of hyperbaric oxygen on vascular endothelial function in patients with slow coronary flow. Cardiol. J. 2018, 25, 106–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Uusijärvi, J.; Eriksson, K.; Larsson, A.C.; Nihlén, C.; Schiffer, T.; Lindholm, P.; Weitzberg, E. Effects of hyperbaric oxygen on nitric oxide generation in humans. Nitric Oxide 2015, 44, 88–97. [Google Scholar] [CrossRef]
  62. Sureda, A.; Batle, J.M.; Martorell, M.; Capó, X.; Tejada, S.; Tur, J.A.; Pons, A. Antioxidant response of chronic wounds to hyperbaric oxygen therapy. PLoS ONE 2016, 11, e0163371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gasier, H.G.; Fothergill, D.M. Oxidative stress, antioxidant defenses and nitric oxide production following hyperoxic exposures. Undersea Hyperb. Med. 2013, 40, 125–134. [Google Scholar]
  64. Lambrechts, K.; Pontier, J.-M.; Mazur, A.; Buzzacott, P.; Morin, J.; Wang, Q.; Théron, M.; Guerrero, F. Effect of decompression-induced bubble formation on highly trained divers microvascular function. Physiol. Rep. 2013, 1, e00142. [Google Scholar] [CrossRef] [Green Version]
  65. Dragic, S.; Momcicevic, D.; Zlojutro, B.; Jandric, M.; Kovacevic, T.; Djajić, V.; Gajić, A.; Talić, G.; Kovacevic, P. Serum levels of nitric oxide and endothelin-1 in vasculopathy managed with hyperbaric oxygen therapy. Clin. Hemorheol. Microcirc. 2020, 75, 233–241. [Google Scholar] [CrossRef]
  66. Shaw, F.L.; Winyard, P.; Smerdon, G.R.; Bryson, P.J.; Moody, J.; Eggleton, P. Hyperbaric oxygen treatment induces platelet aggregation and protein release, without altering expression of activation molecules. Clin. Biochem. 2009, 42, 467–476. [Google Scholar] [CrossRef]
  67. Chen, S.-J.; Yu, C.-T.; Cheng, Y.-L.; Yu, S.-Y.; Lo, H.-C. Effects of hyperbaric oxygen therapy on circulating interleukin-8, nitric oxide, and insulin-like growth factors in patients with type 2 diabetes mellitus. Clin. Biochem. 2007, 40, 30–36. [Google Scholar] [CrossRef]
  68. Gurdol, F.; Cimsit, M.; Oner-Iyidogan, Y.; Kocak, H.; Sengun, S.; Yalcinkaya-Demirsoz, S. Collagen synthesis, nitric oxide and asymmetric dimethylarginine in diabetic subjects undergoing hyperbaric oxygen therapy. Physiol. Res. 2010, 59, 423–429. [Google Scholar] [CrossRef] [PubMed]
  69. Puthucheary, Z.A.; Liu, J.; Bennett, M.; Trytko, B.; Chow, S.; Thomas, P.S. Exhaled nitric oxide is decreased by exposure to the hyperbaric oxygen therapy environment. Mediat. Inflamm. 2006, 2006, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Taraldsøy, T.; Bolann, B.J.; Thorsen, E. Reduced nitric oxide concentration in exhaled gas after exposure to hyperbaric hyperoxia. Undersea Hyperb. Med. 2007, 34, 321–327. [Google Scholar] [PubMed]
  71. Niu, C.-C.; Lin, S.-S.; Yuan, L.-J.; Chen, L.-H.; Wang, I.-C.; Tsai, T.-T.; Lai, P.-L.; Chen, W.-J. Hyperbaric oxygen treatment suppresses MAPK signaling and mitochondrial apoptotic pathway in degenerated human intervertebral disc cells. J. Orthop. Res. 2012, 31, 204–209. [Google Scholar] [CrossRef]
  72. Niu, C.-C.; Yuan, L.-J.; Chen, L.-H.; Lin, S.-S.; Tsai, T.-T.; Liao, J.-C.; Lai, P.-L.; Chen, W.-J. Beneficial effects of hyperbaric oxygen on human degenerated intervertebral disk cells via suppression of IL-1β and p38 MAPK signal. J. Orthop. Res. 2010, 29, 14–19. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, I.-C.; Ueng, S.W.N.; Lin, S.-S.; Niu, C.-C.; Yuan, L.-J.; Su, C.-I.; Chen, C.-H.; Chen, W.-J. Effect of Hyperbaric Oxygenation on intervertebral disc degeneration. Spine 2011, 36, 1925–1931. [Google Scholar] [CrossRef] [Green Version]
  74. Gronow, G.; Kähler, W.; Koch, A.; Klause, N. Benzoate hydroxylation. Oxygen Transport to Tissue XXVI 2006, 566, 223–229. [Google Scholar] [CrossRef]
  75. Kähler, W.; Koch, I.; Wohlrab, C.; Kowalski, J.; Witte, J.; Koch, A. Influence of hyperoxia and physical exercise on *OH-radical stress in humans as measured by dihydroxylated benzoates (DHB) in urine. Undersea Hyperb. Med. 2013, 40, 231–238. [Google Scholar]
  76. Muth, C.M.; Glenz, Y.; Klaus, M.; Radermacher, P.; Speit, G.; Leverve, X. Influence of an orally effective SOD on hyperbaric oxygen-related cell damage. Free. Radic. Res. 2004, 38, 927–932. [Google Scholar] [CrossRef] [PubMed]
  77. Gürdöl, F.; Cimşit, M.; Öner-Iyidoğan, Y.; Körpinar, Ş.; Yalçinkaya, S.; Kocak, H. Early and late effects of hyperbaric oxygen treatment on oxidative stress parameters in diabetic patients. Physiol. Res. 2008, 57, 41–47. [Google Scholar] [CrossRef]
  78. Corcoran, T.; Ting, S.; Mas, E.; Phillips, M.; O’Loughlin, E.; Barden, A.; Mori, T.A. Hyperbaric oxygen therapy is not associated with oxidative stress assessed using plasma F2-isoprostanes and isofurans. Prostaglandins, Leukot. Essent. Fat. Acids 2017, 127, 16–19. [Google Scholar] [CrossRef]
  79. Rockswold, S.B.; Rockswold, G.L.; Zaun, D.A.; Zhang, X.; Cerra, C.E.; Bergman, T.A.; Liu, J. A prospective, randomized clinical trial to compare the effect of hyperbaric to normobaric hyperoxia on cerebral metabolism, intracranial pressure, and oxygen toxicity in severe traumatic brain injury. J. Neurosurg. 2010, 112, 1080–1094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Benedetti, S.; Lamorgese, A.; Piersantelli, M.; Pagliarani, S.; Benvenuti, F.; Canestrari, F. Oxidative stress and antioxidant status in patients undergoing prolonged exposure to hyperbaric oxygen. Clin. Biochem. 2004, 37, 312–317. [Google Scholar] [CrossRef]
  81. Ma, L.; Li, P.; Shi, Z.; Hou, T.; Chen, X.; Du, J. A prospective, randomized, controlled study of hyperbaric oxygen therapy: Effects on healing and oxidative stress of ulcer tissue in patients with a diabetic foot ulcer. Ostomy Wound Manag. 2013, 59, 18–24. [Google Scholar]
  82. Matzi, V.; Greilberger, J.F.; Lindenmann, J.; Neuboeck, N.; Nuhsbaumer, S.; Zelzer, S.; Tafeit, E.; Maier, A.; Smolle-Juettner, F.-M. Application of hyperbaric oxygen reduce oxidative damage of plasmatic carbonyl proteins and 8-OHdG by activating glutathion peroxidase. Clin. Lab. 2015, 61, 587–593. [Google Scholar] [CrossRef]
  83. TekSam, O.; Sabuncuoğlu, S.; Girgin, G.; Özgüneş, H. Evaluation of oxidative stress and antioxidant parameters in children with carbon monoxide poisoning. Hum. Exp. Toxicol. 2019, 38, 1235–1243. [Google Scholar] [CrossRef] [PubMed]
  84. Li, N.; Meng, X.E.; Guo, D.Z.; Fan, D.F.; Pan, S.Y. Wound healing process and related laboratory indexes in patients with type 2 diabetes mellitus after hyperbaric oxygen intervention. Biomed. Res. India 2017, 28, 8838–8843. [Google Scholar]
  85. Paprocki, J.; Sutkowy, P.; Piechocki, J.; Woźniak, A. Markers of oxidant-antioxidant equilibrium in patients with sudden sensorineural hearing loss treated with hyperbaric oxygen therapy. Oxidative Med. Cell. Longev. 2019, 2019, 1–8. [Google Scholar] [CrossRef] [PubMed]
  86. Bearden, S.E.; Cheuvront, S.N.; Ringes, E.M. Oxidative stress during a 3.5-hour exposure to 120 kPa(a) PO2 in human divers. Undersea Hyperb. Med. 1999, 26, 159–164. [Google Scholar] [PubMed]
  87. Rocco, M.; Antonelli, M.; Letizia, V.; Alampi, D.; Spadetta, G.; Passariello, M.; Conti, G.; Serio, P.; Gasparetto, A. Lipid peroxidation, circulating cytokine and endothelin 1 levels in healthy volunteers undergoing hyperbaric oxygenation. Minerva Anestesiol. 2001, 67, 393–400. [Google Scholar] [PubMed]
  88. Chen, C.-Y.; Wu, R.-W.; Tsai, N.-W.; Lee, M.S.; Lin, W.-C.; Hsu, M.-C.; Huang, C.-C.; Lai, Y.-R.; Kung, C.-T.; Wang, H.-C.; et al. Increased circulating endothelial progenitor cells and improved short-term outcomes in acute non-cardioembolic stroke after hyperbaric oxygen therapy. J. Transl. Med. 2018, 16, 255. [Google Scholar] [CrossRef] [Green Version]
  89. Paprocki, J.; Pawłowska, M.; Sutkowy, P.; Piechocki, J.; Woźniak, A. evaluation of oxidative stress in patients with difficult-to-heal skin wounds treated with hyperbaric oxygen. Oxidative Med. Cell. Longev. 2020, 2020, 1–8. [Google Scholar] [CrossRef]
  90. Taylor, L.; Midgley, A.W.; Sandstrom, M.E.; Chrismas, B.; McNaughton, L.R. The effect of the hyperbaric environment on heat shock protein 72 expression in vivo. Res. Sports Med. 2012, 20, 142–153. [Google Scholar] [CrossRef]
  91. Burgos, C.; Henríquez-Olguín, C.; Andrade, D.C.; Ramírez-Campillo, R.; Araneda, O.F.; White, A.; Cerda-Kohler, H. Effects of exercise training under hyperbaric oxygen on oxidative stress markers and endurance performance in young soccer players: A pilot study. J. Nutr. Metab. 2016, 2016, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Dise, C.A.; Clark, J.M.; Lambersten, C.J.; Goodman, D.B. Hyperbaric hyperoxia reversibly inhibits erythrocyte phospholipid fatty acid turnover. J. Appl. Physiol. 1987, 62, 533–538. [Google Scholar] [CrossRef]
  93. Kot, J.; Sićko, Z.; Woźniak, M. Oxidative stress during oxygen tolerance test. Int. Marit. Health 2003, 54, 117–126. [Google Scholar]
  94. Dennog, C.; Radermacher, P.; Barnett, Y.; Speit, G. Antioxidant status in humans after exposure to hyperbaric oxygen. Mutat. Res. Mol. Mech. Mutagen. 1999, 428, 83–89. [Google Scholar] [CrossRef]
  95. Rothfuss, A.; Radermacher, P.; Speit, G. Involvement of heme oxygenase-1 (HO-1) in the adaptive protection of human lymphocytes after hyperbaric oxygen (HBO) treatment. Carcinogenesis 2001, 22, 1979–1985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rothfuss, A.; Speit, G. Investigations on the mechanism of hyperbaric oxygen (HBO)-induced adaptive protection against oxidative stress. Mutat. Res. Mol. Mech. Mutagen. 2002, 508, 157–165. [Google Scholar] [CrossRef]
  97. Speit, G.; Dennog, C.; Eichhorn, U.; Rothfuβ, A.; Kaina, B. Induction of heme oxygenase-1 and adaptive protection against the induction of DNA damage after hyperbaric oxygen treatment. Carcinogenesis 2000, 21, 1795–1799. [Google Scholar] [CrossRef] [Green Version]
  98. Tillmans, F.; Sharghi, R.; Noy, T.; Kähler, W.; Klapa, S.; Sartisohn, S.; Sebens, S.; Koch, A. Effect of hyperoxia on the immune status of oxygen divers and endurance athletes. Free. Radic. Res. 2019, 53, 522–534. [Google Scholar] [CrossRef]
  99. Handy, R.D.; Bryson, P.; Moody, A.J.; Handy, L.M.; Sneyd, J.R. Oxidative metabolism in platelets, platelet aggregation, and hematology in patients undergoing multiple hyperbaric oxygen exposures. Undersea Hyperb. Med. 2006, 32, 327–340. [Google Scholar]
  100. Ansari, K.; Wilson, M.; Slater, G.; Haglin, J.; Kaplan, E. Hyperbaric oxygenation and erythrocyte antioxidant enzymes in multiple sclerosis patients. Acta Neurol. Scand. 1986, 74, 156–160. [Google Scholar] [CrossRef]
  101. Rossignol, D.A.; Rossignol, L.W.; James, S.J.; Melnyk, S.; Mumper, E. The effects of hyperbaric oxygen therapy on oxidative stress, inflammation, and symptoms in children with autism: An open-label pilot study. BMC Pediatr. 2007, 7, 36. [Google Scholar] [CrossRef] [Green Version]
  102. Chang, J.S.; Chang, E.; Lee, Y.; Cha, Y.S.; Cha, S.-K.; Gil Cho, W.; Jeong, Y.; Kim, H.; Park, K.-S. Hyperbaric oxygen exposure attenuates circulating stress biomarkers: A pilot interventional study. Int. J. Environ. Res. Public Health 2020, 17, 7853. [Google Scholar] [CrossRef] [PubMed]
  103. Karadurmus, N.; Sahin, M.; Tasci, C.; Naharci, I.; Ozturk, C.; Ilbasmis, S.; Dulkadir, Z.; Sen, A.; Sağlam, K. Potential benefits of hyperbaric oxygen therapy on atherosclerosis and glycaemic control in patients with diabetic foot. Endokrynol. Pol. 2010, 61, 275–279. [Google Scholar] [PubMed]
  104. Akcali, G.; Uzun, G.; Arziman, I.; Aydin, I.; Yildiz, S. The relationship between intoxication severity and blood interleukin 6, interleukin 10 and CRP levels in carbon monoxide-poisoned patients. Undersea Hyperb. Med. 2019, 45, 646–652. [Google Scholar]
  105. Vezzani, G.; Socias, S.; Bianco, A.; Paoli, A.; Caberti, L.; Cantadori, L.; Manelli, D.; Mordacci, M.; Mangar, D.; Camporesi, E.M.; et al. Inflammatory mediators and other biomarkers in co-intoxicated patients after hyperbaric oxygen therapy (HBO2). Acta Med. Mediterr. 2016, 32, 189–193. [Google Scholar] [CrossRef]
  106. Chen, C.-Y.; Wu, R.-W.; Hsu, M.-C.; Hsieh, C.-J.; Chou, M.-C. Adjunctive hyperbaric oxygen therapy for healing of chronic diabetic foot ulcers. J. Wound Ostomy Cont. Nurs. 2017, 44, 536–545. [Google Scholar] [CrossRef]
  107. Top, C.; Yildiz, S.; Öncül, O.; Qydedi, T.; Çevikbaş, A.; Soyogul, U.G.; Çavuşlu, S. Phagocytic activity of neutrophils improves over the course of therapy of diabetic foot infections. J. Infect. 2007, 55, 369–373. [Google Scholar] [CrossRef]
  108. Yildiz, H.; Senol, L.; Ercan, E.; Bilgili, M.E.; Abuaf, O.K. A prospective randomized controlled trial assessing the efficacy of adjunctive hyperbaric oxygen therapy in the treatment of hidradenitis suppurativa. Int. J. Dermatol. 2015, 55, 232–237. [Google Scholar] [CrossRef] [PubMed]
  109. Xie, Z.; Zhuang, M.; Lin, L.; Xu, H.; Chen, L.; Hu, L. Changes of plasma C-reactive protein in patients with craniocerebral injury before and after hyperbaric oxygenation: A randomly controlled study. Neural Regen. Res. 2007, 2, 314–317. [Google Scholar]
  110. Schnittger, V.; Rosendahl, K.; Lind, F.; Palmblad, J. Effects of carbon monoxide poisoning on neutrophil responses in patients treated with hyperbaric oxygen. J. Investig. Med. 2004, 52, 523–530. [Google Scholar] [CrossRef]
  111. Hao, Y.; Dong, X.; Zhang, M.; Liu, H.; Zhu, L.; Wang, Y. Effects of hyperbaric oxygen therapy on the expression levels of the inflammatory factors interleukin-12p40, macrophage inflammatory protein-1β, platelet-derived growth factor-BB, and interleukin-1 receptor antagonist in keloids. Med. Baltim. 2020, 99, e19857. [Google Scholar] [CrossRef]
  112. Bent, S.; Bertoglio, K.; Ashwood, P.; Nemeth, E.; Hendren, R.L. Brief report: Hyperbaric oxygen therapy (HBOT) in children with autism spectrum disorder: A clinical trial. J. Autism Dev. Disord. 2011, 42, 1127–1132. [Google Scholar] [CrossRef]
  113. Aydin, F.; Kaya, A.; Karapinar, L.; Kumbaraci, M.; Imerci, A.; Karapinar, H.; Karakuzu, C.; Incesu, M. IGF-1 Increases with hyperbaric oxygen therapy and promotes wound healing in diabetic foot ulcers. J. Diabetes Res. 2013, 2013, 1–6. [Google Scholar] [CrossRef] [PubMed]
  114. Hedetoft, M.; Olsen, N.V.; Smidt-Nielsen, I.G.; Wahl, A.M.; Bergström, A.; Juul, A.; Hyldegaard, O. Measurement of peripheral arterial tonometry in patients with diabetic foot ulcers during courses of hyperbaric oxygen treatment. Diving Hyperb. Med. J. 2020, 50, 17–23. [Google Scholar] [CrossRef] [PubMed]
  115. Romero-Valdovinos, M.; Cárdenas-Mejía, A.; Gutiérrez-Gómez, C.; Flisser, A.; Kawa-Karasik, S.; Ortiz-Monasterio, F. Keloid skin scars: The influence of hyperbaric oxygenation on fibroblast growth and on the expression of messenger RNA for insulin like growth factor and for transforming growth factor. Vitr. Cell. Dev. Biol. Anim. 2011, 47, 421–424. [Google Scholar] [CrossRef] [PubMed]
  116. Irawan, H.; Semadi, I.N.; Devi, A. Effect of hyperbaric oxygen therapy to improve serum albumin for patients with diabetic foot ulcers. Biomed. Pharmacol. J. 2018, 11, 569–575. [Google Scholar] [CrossRef]
  117. Irawan, H.; Semadi, I.N.; Widiana, I.G.R. A pilot study of short-duration hyperbaric oxygen therapy to improve HbA1c, Leukocyte, and serum creatinine in patients with diabetic foot ulcer Wagner 3–4. Sci. World J. 2018, 2018, 1–6. [Google Scholar] [CrossRef] [Green Version]
  118. Chong, S.J.; Kan, E.M.; Song, C.; Soh, C.R.; Lu, J. Characterization of early thermal burns and the effects of hyperbaric oxygen treatment: A pilot study. Diving Hyperb. Med. J. 2013, 43, 157–161. [Google Scholar]
  119. Sun, J.; Zheng, J.; Wang, F.; Zhang, G.; Wu, J. Effect of hyperbaric oxygen combined with nimodipine on treatment of diffuse brain injury. Exp. Ther. Med. 2018, 15, 4651–4658. [Google Scholar] [CrossRef] [Green Version]
  120. Baiula, M.; Greco, R.; Ferrazzano, L.; Caligiana, A.; Hoxha, K.; Bandini, D.; Longobardi, P.; Spampinato, S.; Tolomelli, A. Integrin-mediated adhesive properties of neutrophils are reduced by hyperbaric oxygen therapy in patients with chronic non-healing wound. PLoS ONE 2020, 15, e0237746. [Google Scholar] [CrossRef]
  121. Benson, R.M.; Minter, L.M.; Osborne, B.A.; Granowitz, E.V. Hyperbaric oxygen inhibits stimulus-induced proinflammatory cytokine synthesis by human blood-derived monocyte-macrophages. Clin. Exp. Immunol. 2003, 134, 57–62. [Google Scholar] [CrossRef] [Green Version]
  122. Fan, A.; Zhou, J. Effect of the combination of donepezil with hyperbaric oxygen therapy and functional rehabilitation training on parkinson’s disease dementia and the neurological function system. Int. J. Clin. Exp. Med. 2020, 13, 5867–5875. [Google Scholar]
  123. Wilkinson, D.; Nolting, M.; Mahadi, M.K.; Chapman, I.; Heilbronn, L. Hyperbaric oxygen therapy increases insulin sensitivity in overweight men with and without type 2 diabetes. Diving Hyperb. Med. J. 2015, 45, 30–36. [Google Scholar]
  124. Weisz, G.; Lavy, A.; Adir, Y.; Melamed, Y.; Rubin, D.; Eidelman, S.; Pollack, S. Modification of in vivo and in vitro TNF-α, IL-1, and IL-6 secretion by circulating monocytes during hyperbaric oxygen treatment in patients with perianal crohn’s disease. J. Clin. Immunol. 1997, 17, 154–159. [Google Scholar] [CrossRef]
  125. Anguiano-Hernandez, Y.M.; Contreras-Mendez, L.; Hernandez-Cueto, M.D.L.A.; Muñoz-Medina, J.E.; Santillan-Verde, M.A.; Barbosa-Cabrera, R.E.; Delgado-Quintana, C.A.; Trejo-Rosas, S.; Santacruz-Tinoco, C.E.; Gonzalez-Ibarra, J.; et al. Modification of HIF-1α, NF-κB, IGFBP-3, VEGF and adiponectin in diabetic foot ulcers treated with hyperbaric oxygen. Undersea Hyperb. Med. 2019, 46, 35–44. [Google Scholar] [CrossRef]
  126. MacKenzie, D.A.; Sollinger, H.W.; Hullett, D.A. Role of CD4+ regulatory T cells in hyperbaric oxygen-mediated immune nonresponsiveness. Hum. Immunol. 2000, 61, 1320–1331. [Google Scholar] [CrossRef]
  127. Alex, J.; Laden, G.; Cale, A.R.; Bennett, S.; Flowers, K.; Madden, L.; Gardiner, E.; McCollum, P.T.; Griffin, S.C. Pretreatment with hyperbaric oxygen and its effect on neuropsychometric dysfunction and systemic inflammatory response after cardiopulmonary bypass: A prospective randomized double-blind trial. J. Thorac. Cardiovasc. Surg. 2005, 130, 1623–1630. [Google Scholar] [CrossRef] [Green Version]
  128. Fildissis, G.; Venetsanou, K.; Myrianthefs, P.; Karatzas, S.; Zidianakis, V.; Baltopoulos, G. Whole blood pro-inflammatory cytokines and adhesion molecules post-lipopolysaccharides exposure in hyperbaric conditions. Eur. Cytokine Netw. 2004, 15, 217–221. [Google Scholar]
  129. Hou, S.; Wu, G.; Liang, J.; Cheng, H.; Chen, C. Hyperbaric oxygen on rehabilitation of brain tumors after surgery and effects on TNF-α and IL-6 levels. Oncol. Lett. 2019, 17, 3277–3282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Lin, P.-Y.; Sung, P.-H.; Chung, S.-Y.; Hsu, S.-L.; Chung, W.-J.; Sheu, J.-J.; Hsueh, S.-K.; Chen, K.-H.; Wu, R.-W.; Yip, H.-K. Hyperbaric oxygen therapy enhanced circulating levels of endothelial progenitor cells and angiogenesis biomarkers, blood flow, in ischemic areas in patients with peripheral arterial occlusive disease. J. Clin. Med. 2018, 7, 548. [Google Scholar] [CrossRef] [Green Version]
  131. Rosario, E.R.; Kaplan, S.E.; Khonsari, S.; Vazquez, G.; Solanki, N.; Lane, M.; Brownell, H.; Rosenberg, S.S. The effect of hyperbaric oxygen therapy on functional impairments caused by ischemic stroke. Neurol. Res. Int. 2018, 2018, 1–12. [Google Scholar] [CrossRef] [Green Version]
  132. Song, K.-X.; Liu, S.; Zhang, M.-Z.; Liang, W.-Z.; Liu, H.; Dong, X.-H.; Wang, Y.-B.; Wang, X.-J. Hyperbaric oxygen therapy improves the effect of keloid surgery and radiotherapy by reducing the recurrence rate. J. Zhejiang Univ. Sci. B 2018, 19, 853–862. [Google Scholar] [CrossRef]
  133. Guggino, G.; Schinocca, C.; Pizzo, M.L.; Di Liberto, D.; Garbo, D.; Raimondo, S.; Alessandro, R.; Brighina, F.; Ruscitti, P.; Giacomelli, R.; et al. T helper 1 response is correlated with widespread pain, fatigue, sleeping disorders and the quality of life in patients with fibromyalgia and is modulated by hyperbaric oxygen therapy. Clin. Exp. Rheumatol. 2019, 37, 81–89. [Google Scholar]
  134. Wang, B.-W.; Lin, C.-M.; Wu, G.-J.; Shyu, K.-G. Tumor necrosis factor-α enhances hyperbaric oxygen-induced visfatin expression via JNK pathway in human coronary arterial endothelial cells. J. Biomed. Sci. 2011, 18, 27. [Google Scholar] [CrossRef] [Green Version]
  135. Nasole, E.; Nicoletti, C.; Yang, Z.-J.; Girelli, A.; Rubini, A.; Giuffreda, F.; Di Tano, A.; Camporesi, E.; Bosco, G. Effects of alpha lipoic acid and its R+ enantiomer supplemented to hyperbaric oxygen therapy on interleukin-6, TNF-α and EGF production in chronic leg wound healing. J. Enzym. Inhib. Med. Chem. 2013, 29, 297–302. [Google Scholar] [CrossRef]
  136. Yoshinoya, Y.; Böcker, A.H.; Ruhl, T.; Siekmann, U.; Pallua, N.; Beier, J.P.; Kim, B.-S. The effect of hyperbaric oxygen therapy on human adipose-derived stem cells. Plast. Reconstr. Surg. 2020, 146, 309–320. [Google Scholar] [CrossRef]
  137. Liu, X.H.; Liang, F.; Jia, X.Y.; Zhao, L.; Zhou, Y.; Yang, J. Hyperbaric oxygen treatment improves hearing level via attenuating TLR4/NF-κB mediated inflammation in Sudden sensorineural hearing loss patients. Biomed. Environ. Sci. 2020, 33, 331–337. [Google Scholar] [CrossRef]
  138. Semadi, N.I. The role of VEGF and TNF-Alpha on epithelialization of diabetic foot ulcers after hyperbaric oxygen therapy. Open Access Maced. J. Med Sci. 2019, 7, 3177–3183. [Google Scholar] [CrossRef] [Green Version]
  139. Madden, L.A.; Vince, R.V.; Laden, G. The effect of acute hyperoxia in vivo on NF kappa B expression in human PBMC. Cell Biochem. Funct. 2010, 29, 71–73. [Google Scholar] [CrossRef]
  140. Sun, L.; Zhao, L.; Li, P.; Liu, X.; Liang, F.; Jiang, Y.; Kang, N.; Gao, C.; Yang, J. Effect of hyperbaric oxygen therapy on HMGB1/NF-κB expression and prognosis of acute spinal cord injury: A randomized clinical trial. Neurosci. Lett. 2019, 692, 47–52. [Google Scholar] [CrossRef]
  141. Tra, W.M.W.; Spiegelberg, L.; Tuk, B.; Hovius, S.E.R.; Perez-Amodio, S. Hyperbaric oxygen treatment of tissue-engineered mucosa enhances secretion of angiogenic factors in vitro. Tissue Eng. Part A 2014, 20, 1523–1530. [Google Scholar] [CrossRef]
  142. Lee, C.-C.; Chen, S.-C.; Tsai, S.-C.; Wang, B.-W.; Liu, Y.-C.; Lee, H.-M.; Shyu, K.-G. Hyperbaric oxygen induces VEGF expression through ERK, JNK and c-Jun/AP-1 activation in human umbilical vein endothelial cells. J. Biomed. Sci. 2005, 13, 143–156. [Google Scholar] [CrossRef] [Green Version]
  143. Jung, S.; Wermker, K.; Poetschik, H.; Ziebura, T.; Kleinheinz, J. The impact of hyperbaric oxygen therapy on serological values of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Head Face Med. 2010, 6, 29. [Google Scholar] [CrossRef] [Green Version]
  144. Lin, S.; Shyu, K.-G.; Lee, C.-C.; Wang, B.-W.; Chang, C.-C.; Liu, Y.-C.; Huang, F.-Y.; Chang, H. Hyperbaric oxygen selectively induces angiopoietin-2 in human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun. 2002, 296, 710–715. [Google Scholar] [CrossRef]
  145. Mutzbauer, T.; Schneider, M.; Neubauer, B.; Weiss, M.; Tetzlaff, K. Antioxidants may attenuate plasma erythropoietin decline after hyperbaric oxygen diving. Int. J. Sports Med. 2015, 36, 1035–1040. [Google Scholar] [CrossRef]
  146. Wu, L.L.; Chiou, C.-C.; Chang, P.-Y.; Wu, J.T. Urinary 8-OHdG: A marker of oxidative stress to DNA and a risk factor for cancer, atherosclerosis and diabetics. Clin. Chim. Acta 2004, 339, 1–9. [Google Scholar] [CrossRef]
  147. Kang, T.S.; Gorti, G.K.; Quan, S.Y.; Ho, M.; Koch, R.J. Effect of hyperbaric oxygen on the growth factor profile of fibroblasts. Arch. Facial Plast. Surg. 2004, 6, 31–35. [Google Scholar] [CrossRef] [Green Version]
  148. Kunnavatana, S.S.; Quan, S.Y.; Koch, R.J. Combined effect of hyberbaric oxygen and n-acetylcysteine on fibroblast proliferation. Arch. Otolaryngol. Head Neck Surg. 2005, 131, 809–814. [Google Scholar] [CrossRef] [Green Version]
  149. Naito, Y.; Lee, M.; Kato, Y.; Nagai, R.; Yonei, Y. Oxidative Stress Markers. Anti-Aging Med. 2010, 7, 36–44. [Google Scholar] [CrossRef] [Green Version]
  150. Gabay, C.; Kushner, I. Acute-phase proteins and other systemic responses to inflammation. N. Engl. J. Med. 1999, 340, 448–454. [Google Scholar] [CrossRef]
  151. Ansar, W.; Ghosh, S. Inflammation and Inflammatory Diseases, Markers, and Mediators: Role of CRP in Some Inflammatory Diseases. In Biology of C Reactive Protein in Health and Disease; Springer: New Delhi, India, 2016; pp. 67–107. [Google Scholar]
  152. Biswas, S.K. Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxidative Med. Cell. Longev. 2016, 2016, 1–9. [Google Scholar] [CrossRef] [Green Version]
  153. Vaziri, N.D.; Rodríguez-Iturbe, B. Mechanisms of disease: Oxidative stress and inflammation in the pathogenesis of hypertension. Nat. Clin. Pract. Nephrol. 2006, 2, 582–593. [Google Scholar] [CrossRef]
  154. Chatterjee, S. Oxidative Stress, Inflammation, and Disease. In Oxidative Stress and Biomaterials; Academic Press: Cambridge, MA, USA, 2016; pp. 35–58. [Google Scholar]
  155. Glik, J.; Cholewka, A.; Stanek, A.; Sieroń, K.; Mikuś-Zagórska, K.; Knefel, G.; Nowak, M.; Kawecki, M.; Englisz, B. Thermal imaging and planimetry evaluation of the results of chronic wounds treatment with hyperbaric oxygen therapy. Adv. Clin. Exp. Med. 2019, 28, 229–236. [Google Scholar] [CrossRef]
  156. Englisz-Jurgielewicz, B.; Cholewka, A.; Firganek, E.; Knefel, G.; Kawecki, M.; Glik, J.; Nowak, M.; Sieroń, K.; Stanek, A. Evaluation of hyperbaric oxygen therapy effects in hard-to-heal wounds using thermal imaging and planimetry. J. Therm. Anal. Calorim. 2019, 141, 1465–1475. [Google Scholar] [CrossRef]
  157. Frangogiannis, N.; Smith, C.; Entman, M.L. The inflammatory response in myocardial infarction. Cardiovasc. Res. 2002, 53, 31–47. [Google Scholar] [CrossRef]
  158. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [Green Version]
  159. Lawrence, T. The nuclear factor NF- B pathway in inflammation. Cold Spring Harb. Perspect. Biol. 2009, 1, a001651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Yu, X.; Li, Y.; He, X.; Li, X.; Din, B.; Gan, Y.; Xu, M. Hyperbaric oxygen reduces inflammatory response in acute pancreatitis by inhibiting NF-κB activation. Eur. Surg. Res. 2009, 42, 130–135. [Google Scholar] [CrossRef]
  161. Szade, A.; Grochot-Przeczek, A.; Florczyk, U.; Jozkowicz, A.; Dulak, J. Cellular and molecular mechanisms of inflammation-induced angiogenesis. IUBMB Life 2015, 67, 145–159. [Google Scholar] [CrossRef]
  162. Fallah, A.; Sadeghinia, A.; Kahroba, H.; Samadi, A.; Heidari, H.R.; Bradaran, B.; Zeinali, S.; Molavi, O. Therapeutic targeting of angiogenesis molecular pathways in angiogenesis-dependent diseases. Biomed. Pharmacother. 2019, 110, 775–785. [Google Scholar] [CrossRef]
  163. Maulik, N. Redox signaling of angiogenesis. Antioxid. Redox Signal. 2002, 4, 805–815. [Google Scholar] [CrossRef]
  164. Dejmek, J.; Kohoutová, M.; Kripnerová, M.; Cedikova, M.; Tůma, Z.; Babuška, V.; Bolek, L.; Kuncová, J. repeated exposure to hyperbaric hyperoxia affects mitochondrial functions of the lung fibroblasts. Physiol. Res. 2018, 67, S633–S643. [Google Scholar] [CrossRef] [PubMed]
  165. Sinan, M.; Ertan, N.Z.; Mirasoglu, B.; Yalcin, O.; Atac, N.; Toklu, A.S.; Basaran-Kucukgergin, C.; Baskurt, O.K. Acute and long-term effects of hyperbaric oxygen therapy on hemorheological parameters in patients with various disorders. Clin. Hemorheol. Microcirc. 2016, 62, 79–88. [Google Scholar] [CrossRef] [PubMed]
Figure 1. PRISMA Flow Diagram of the selection progress.
Figure 1. PRISMA Flow Diagram of the selection progress.
Biomolecules 11 01210 g001
Table 1. The effect of HBOT on oxidative stress markers.
Table 1. The effect of HBOT on oxidative stress markers.
Main AspectAssociated MarkersStimulating
Effect
No EffectInhibiting Effect
Causers of
oxidative stress
Reactive oxygen species (including superoxide-ion and hydrogen peroxide)[33,34,35,36,37,38,39,40,41][33,37,38,42,43][33,42,43,44]
Nitric oxide synthase (NOS) (including endothelial NOS and inducible NOS)[34,45,46,47,48][49,50,51][52,53,54]
Reactive nitrogen species (including nitric oxygen, nitrite and nitrate)[33,34,45,49,55,56,57,58,59,60][33,46,61,62,63,64,65,66,67,68][53,61,67,68,69,70,71,72,73]
Hydrobenzoates[74,75][74]
Free fatty acid [53]
Myeloperoxidase [34,62]
Lipid peroxidationIsoprostanes[76,77][78,79]
Isofurans [78]
Malondialdehyde[56,77,80,81,82][14,34,49,55,76,83][62,84]
Thiobarbituric acid reactive substances[63,74,85,86,87][33,63,85,88,89,90]
Lipid hydroperoxides[36][91][92]
Oxidized low-density lipoprotein [82]
Protein
peroxidation
Nitrotyrosine[49][64]
Advanced oxidation protein products [77]
Carbohydrate
peroxidation
Protein carbonyls[93]
Carbonyl group[56]
Protein carbonyl derivates [55]
Plasma carbonyl proteins [83][82]
DNA/RNA
damage
8-hydroxydeoxyguanosine [83][82]
Tail moment[38,94,95,96][76,97,98]
Sister chromatid exchange[14]
Gene expressionNuclear factor erythroid 2- related factor 2[45]
Other residues of oxidative stressReactive oxygen metabolites[80]
Intracellular calcium concentration [43]
AntioxidantsTotal antioxidant capacity [91,93,94,99]
Catalase[33,34,43,45,55,62,81,100][43,55,62,63,76,83,89,94,97][63,80,85]
Superoxide dismutase[55,56,81,84,100,101][14,33,55,62,63,76,83,89,94,97,102][80,85,86]
Glutathione[92][76,80,83,94]
Glutathione disulfide [76,101]
Glutathione reductase [55,62]
Glutathione peroxidase[34,63,82,85][14,55,62,63,76,80,83,89,94,100][63]
Thiols [88][93]
Vitamin A [80,94]
Vitamin C [94]
Vitamin E [80,94]
Uric acid [91,102][103]
Heme oxygenase-1[45,95,96,97]
NAD(P)H dehydrogenase [quinone] 1[45]
Table 2. The effect of HBOT on inflammation markers.
Table 2. The effect of HBOT on inflammation markers.
Main AspectAssociated MarkersStimulating EffectNo EffectInhibiting Effect
Acute-phase proteins(high-sensitivity) C-reactive protein [88,101,104,105][45,53,58,59,60,84,101,103,106,107,108,109]
Granulocyte-colony stimulating factor[110][110,111,112]
Ferritin [96]
Insulin-like growth factor-1[113][67,114,115][114,115]
Albumin[116][102,117]
Interleukins (IL)IL-1α[45][111,112]
IL-1β [35,87,111,112,118,119][71,72,73,120,121,122]
IL-1Ra[111][123]
IL-1 [4][124]
IL-4 [111,112,118,125,126]
IL-6[62,123,125][87,104,105,111,112,118,123,127,128][4,35,122,124,129,130,131,132]
IL-8[45][4,67,105,110,111,112,128][46,105,127]
IL-10[45][106,107,113,114,120,127,128][4,105]
Interferons (IFN)IFN-α [111,112]
IFN-γ[118][111,112,125,126][45,133]
CytokinesTumor necrosis factor-α[87,127,134][4,45,111,112,128,135,136][35,84,105,119,120,121,123,124,129,131,132,133,137,138]
Nuclear factor kappa B[139][125][52,53,125,132,137,140]
OthersErythrocyte sedimentation rate[107] [106,108]
Table 3. The effect of HBOT on angiogenesis markers.
Table 3. The effect of HBOT on angiogenesis markers.
Main AspectAssociated MarkersStimulating
Effect
No EffectInhibiting Effect
Growth factors/
cytokines
Vascular endothelial growth factor[45,62,84,125,130,138,141,142][50,51,113,120,127,137,143,144,145,146][132]
(basic) Fibroblast growth factor[45,141][102,111,147,148][141,143]
Platelet-derived growth factor [45,111][111]
Insulin-like growth factor-binding protein[125][67,125][67]
Epidermal growth factor[45][50,111,135,136]
Insulin-like growth factor-2 [67]
Hematopoietic growth factor[130,141][136]
Keratinocyte growth factor[141]
Placental growth factor[40,141] [141]
Tumor growth factor-α [111]
Tumor growth factor-β [112,147,148][115,136]
Angiopoietin[84][144]
Erythropoietin [145]
Granulocyte-macrophage colony-stimulating factor [111,112]
Stromal cell-derived factor-1α[130]
Cytokine
receptors
Tie-2 [144]
Erythropoietin-receptor [37]
ProteasesMatrix metalloproteinase-3 [52,54,72]
Matrix metalloproteinase-9 [52]
Matrix metalloproteinase-13 [52]
Transcription factorsHypoxia-inducible factor-1α [125][47,130,132]
Downstream effectorsPhosphatidylinositol-3 kinase (PI3K)[48]
AKT[48] [53]
p38 mitogen-activated protein kinase (p38 MAPK) [44,52,71,72]
Extracellular signal-regulated kinase (ERK)[142][44][52,53,71]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

De Wolde, S.D.; Hulskes, R.H.; Weenink, R.P.; Hollmann, M.W.; Van Hulst, R.A. The Effects of Hyperbaric Oxygenation on Oxidative Stress, Inflammation and Angiogenesis. Biomolecules 2021, 11, 1210. https://doi.org/10.3390/biom11081210

AMA Style

De Wolde SD, Hulskes RH, Weenink RP, Hollmann MW, Van Hulst RA. The Effects of Hyperbaric Oxygenation on Oxidative Stress, Inflammation and Angiogenesis. Biomolecules. 2021; 11(8):1210. https://doi.org/10.3390/biom11081210

Chicago/Turabian Style

De Wolde, Silke D., Rick H. Hulskes, Robert P. Weenink, Markus W. Hollmann, and Robert A. Van Hulst. 2021. "The Effects of Hyperbaric Oxygenation on Oxidative Stress, Inflammation and Angiogenesis" Biomolecules 11, no. 8: 1210. https://doi.org/10.3390/biom11081210

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop