ArticlePDF Available

Abstract and Figures

Introduction Hyperbaric air (HBA) was first used pharmaceutically in 1662 to treat lung disease. Extensive use in Europe and North America followed throughout the 19th century to treat pulmonary and neurological disorders. HBA reached its zenith in the early 20th century when cyanotic, moribund “Spanish flu pandemic” patients turned normal color and regained consciousness within minutes after HBA treatment. Since that time the 78% Nitrogen fraction in HBA has been completely displaced by 100% oxygen to create the modern pharmaceutical hyperbaric oxygen therapy (HBOT), a powerful treatment that is FDA approved for multiple indications. Current belief purports oxygen as the active element mobilizing stem progenitor cells (SPCs) in HBOT, but hyperbaric air, which increases tensions of both oxygen and nitrogen, has been untested until now. In this study we test HBA for SPC mobilization, cytokine and chemokine expression, and complete blood count. Methods Ten 34–35-year-old healthy volunteers were exposed to 1.27ATA (4 psig/965 mmHg) room air for 90 min, M-F, for 10 exposures over 2-weeks. Venous blood samples were taken: (1) prior to the first exposure (served as the control for each subject), (2) directly after the first exposure (to measure the acute effect), (3) immediately prior to the ninth exposure (to measure the chronic effect), and (4) 3 days after the completion of tenth/final exposure (to assess durability). SPCs were gated by blinded scientists using Flow Cytometry. Results SPCs (CD45dim/CD34⁺/CD133⁻) were mobilized by nearly two-fold following 9 exposures (p = 0.02) increasing to three-fold 72-h post completion of the final (10th) exposure (p = 0.008) confirming durability. Discussion This research demonstrates that SPCs are mobilized, and cytokines are modulated by hyperbaric air. HBA likely is a therapeutic treatment. Previously published research using HBA placebos should be re-evaluated to reflect a dose treatment finding rather than finding a placebo effect. Our findings of SPC mobilization by HBA support further investigation into hyperbaric air as a pharmaceutical/therapy.
This content is subject to copyright.
Frontiers in Neurology 01 frontiersin.org
Hyperbaric air mobilizes stem cells
in humans; a new perspective on
the hormetic dose curve
KentJ.MacLaughlin
1
*, GregoryP.Barton
2, RudolfK.Braun
1,
JuliaE.MacLaughlin
3, Jacob J.Lamers
1, Matthew D.Marcou
1
and MarloweW.Eldridge
1
1 Department of Pediatrics, University of Wisconsin–Madison, Madison, WI, United States, 2 Department
of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States,
3 Medical Oxygen Outpatient Clinic, The American Center, Madison, WI, United States
Introduction: Hyperbaric air (HBA) was first used pharmaceutically in 1662 to treat
lung disease. Extensive use in Europe and North America followed throughout the
19th century to treat pulmonary and neurological disorders. HBA reached its zenith
in the early 20th century when cyanotic, moribund “Spanish flu pandemic” patients
turned normal color and regained consciousness within minutes after HBA treatment.
Since that time the 78% Nitrogen fraction in HBA has been completely displaced
by 100% oxygen to create the modern pharmaceutical hyperbaric oxygen therapy
(HBOT), a powerful treatment that is FDA approved for multiple indications. Current
belief purports oxygen as the active element mobilizing stem progenitor cells (SPCs) in
HBOT, but hyperbaric air, which increases tensions of both oxygen and nitrogen, has
been untested until now. In this study wetest HBA for SPC mobilization, cytokine and
chemokine expression, and complete blood count.
Methods: Ten 34–35-year-old healthy volunteers were exposed to 1.27ATA (4
psig/965 mmHg) room air for 90 min, M-F, for 10 exposures over 2-weeks. Venous
blood samples were taken: (1) prior to the first exposure (served as the control for
each subject), (2) directly after the first exposure (to measure the acute eect), (3)
immediately prior to the ninth exposure (to measure the chronic eect), and (4) 3 days
after the completion of tenth/final exposure (to assess durability). SPCs were gated by
blinded scientists using Flow Cytometry.
Results: SPCs (CD45dim/CD34+/CD133-) were mobilized by nearly two-fold following
9 exposures (p = 0.02) increasing to three-fold 72-h post completion of the final (10th)
exposure (p = 0.008) confirming durability.
Discussion: This research demonstrates that SPCs are mobilized, and cytokines are
modulated by hyperbaric air. HBA likely is a therapeutic treatment. Previously published
research using HBA placebos should be re-evaluated to reflect a dose treatment
finding rather than finding a placebo eect. Our findings of SPC mobilization by HBA
support further investigation into hyperbaric air as a pharmaceutical/therapy.
KEYWORDS
CD34+, Traumatic Brain Injury, CD133, war veteran self harm, war veterans’
psychological suering, war veterans suicide, hyperbaric air placebo, HBOT
Introduction
Presently, hyperbaric air (HBA) is not thought of as a medication. Its singular approved
medicinal use is for Acute Mountain Sickness. Historically it has been used medicinally, initially
reported by Henshaw in 1662 to treat “aictions of the lungs” (1) and additional reports from
OPEN ACCESS
EDITED BY
George Mychaskiw,
Ochsner LSU Health, UnitedStates
REVIEWED BY
Ingrid Eftedal,
Norwegian University of Science and
Technology, Norway
Shai Efrati,
Tel Aviv University, Israel
*CORRESPONDENCE
Kent J. MacLaughlin
kmaclaughlin@pediatrics.wisc.edu
RECEIVED 23 March 2023
ACCEPTED 11 May 2023
PUBLISHED 20 June 2023
CITATION
MacLaughlin KJ, Barton GP, Braun RK,
MacLaughlin JE, Lamers JJ, Marcou MD and
Eldridge MW (2023) Hyperbaric air mobilizes
stem cells in humans; a new perspective on the
hormetic dose curve.
Front. Neurol. 14:1192793.
doi: 10.3389/fneur.2023.1192793
COPYRIGHT
© 2023 MacLaughlin, Barton, Braun,
MacLaughlin, Lamers, Marcou and Eldridge.
This is an open-access article distributed under
the terms of the Creative Commons Attribution
License (CC BY). The use, distribution or
reproduction in other forums is permitted,
provided the original author(s) and the
copyright owner(s) are credited and that the
original publication in this journal is cited, in
accordance with accepted academic practice.
No use, distribution or reproduction is
permitted which does not comply with these
terms.
TYPE Original Research
PUBLISHED 20 June 2023
DOI 10.3389/fneur.2023.1192793
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 02 frontiersin.org
Europe and America followed (1). In 1857 Simpson published a paper
using HBA to treat lung pathologies including tuberculosis (2).
Interest in hyperbaric air as a medication surged following successful
treatments of “Spanish u” patients by Cunningham in 1918 (35).
Unfortunately, Cunningham produced a paucity of papers supporting
his work and when hedied in 1937, interest in hyperbaric air abated.
HBA began to be employed again in the modern era with
controversial results. It was used as a placebo by Collet while
investigating the use of hyperbaric oxygen in Cerebral Palsy (6) and
also as a placebo in brain injury by Miller, Wolf and Cifu (79). In
each of these experiments, both the treatment group and the placebo
group improved revealing an apparent placebo eect. e use of
hyperbaric air as a placebo is energetically debated and controversial
because it increases the oxygen tension. In contrast to the placebo
ndings, similar published studies not using the hyperbaric air
placebo, found signicant improvements in brain injury (1020).
Considering the aforementioned, we asked the question, “is
hyperbaric air an appropriate placebo?”. Wesearched the literature
and found no evidence that hyperbaric air has been tested.
Wedesigned a test of hyperbaric air using a gold standard endpoint of
oxygen therapy, stem cell mobilization.
Our specic aims were (1) to characterize stem cell mobilization
in healthy adults following daily exposures to hyperbaric air, (2)
determine if other biomarkers were modulated, (3) determine if there
were acute changes, and (4) if so, were the changes durable. Based on
previous research done in our lab (21) and calculating a 27% increase
in oxygen partial pressures in the inhaled gases, wehypothesized that
(1) stem cells would bemobilized, (2) biomarkers would bemodulated
(3) there would beacute changes and (4) the stem cell mobilization
would bedurable.
Materials and methods
Design and subjects
is prospective hyperbaric air study was a randomized, single-
blind study conducted at the University of Wisconsin – Madison
Clinical Sciences Center between May 1st, 2021, and August 31st,
2021. is study is approved by the Institutional Review Board of the
University of Wisconsin – Madison. UW IRB ID: 2020-0293-CR001.
All participants provided written informed consent. Healthy adults
were recruited for participation in this study (Table1).
Hyperbaric air exposure
All exposures were at 1.27 ATA (4 psig) of room air for 90 min in
a Gamow style Mountain Sickness Chamber (Hyperbaric Technologies
Inc. Amsterdam, NY, UnitedStates). To minimize circadian cell cycle
variations including acrophase and circannual cycle, all subjects were
diurnally active and all hyperbaric exposures and sample collections
occurred at the same time of day over the contiguous
15-day experiment.
Sample collection
Each peripheral venous blood sample was collected during
the subject’s exposure time slot using either a 21- or 23-gauge BD
Vacutainer Safety-Lok Blood Collection Set (Becton, Dickinson
and Company, Franklin Lakes, NJ, UnitedStates) into a Cyto-
Chex BCT tube (Streck Inc. NE USA), BD Vacutainer Plastic
Blood Collection Tubes with K2EDTA, BD Vacutainer Plastic
Blood Collection Tubes – PST Plasma Separation Tubes, and
Greiner Bio-One K2EDTA GelTubes. All samples were stored
according to manufacturer’s directions. Study protocol in graphic
format is included in Figure1.
Flow cytometry
Fluorescence minus one (FMO) tubes, rainbow bead tubes, and
single antibody tubes were prepared as gating references. Antibodies
were pipetted into ow cytometry tubes according to manufacturer’s
instructions. Antibodies used include CD34 = Brilliant Violet 421
(BioLegend, San Diego, CA, UnitedStates), CD45 = Alexa Fluor 488
(BioLegend, San Diego, CA, UnitedStates), CD133 = PE (Miltenyi
Biotec, North Rhine-Westphalia, Germany) CD31 = Brilliant Violet
605 (BioLegend, San Diego, CA, United States), CD105 = PE-Cy7
(BioLegend, San Diego, CA, United States), Ghost Dye Red
780 = Tonbo Biosciences, San Diego, CA.
Flow cytometry was performed by blinded scientists on a
ermoFisher Attune NxT (Waltham, MA, UnitedStates). Samples
were analyzed by blinded scientists using FlowJo soware (FlowJo,
Ashland, OR, UnitedStates).
Enzyme-linked immunosorbent assay
e Invitrogen ProcartaPlex Human Immune Monitoring
Panel 65-Plex (Invitrogen, Waltham, MA, UnitedStates) was used to
assess changes in cytokines, chemokines and growth factors. All tests
were performed by blinded scientists at the University of Wisconsin
Non-Human Primate Research Center (Madison, WI, UnitedStates).
Statistical analysis
e rst blood draw taken prior to the rst exposure served as the
control. To determine whether there was an overall eect across
exposures, weutilized the Friedman test (nonparametric alternative
to one-way ANVOA with repeated measures) and if signicant,
comparisons between all-time points were performed using the
Wilcoxon signed rank test. Signicance level was determined a priori
at the 0.05 level and all tests were two-tailed. Statistical analyses were
calculated using Graph Pad Prism (GraphPad Prism 9.0.0 Soware,
San Diego, CA, UnitedStates).
TABLE1 Anthropometric data of subjects in this hyperbaric air study.
NMean Standard
deviation
Age 10 34.54 years 1.36 years
Height 10 171.75 cm 9.38 cm
Weight 10 81.40 kg 21.05 kg
BMI 10 27.43% 2.8%
Female 5 5 NA
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 03 frontiersin.org
Results
CD45dim
Increased frequency of CD45dim/CD34+/CD133
following nominal exposure to hyperbaric room
air
As previously described 10 humans were exposed to 1.27 ATA of
room air 10 times over the course of a 12-day period. Results revealed
a signicant increase in the frequency of CD45
dim
/CD34
+
/CD133
stem progenitor cells in venous blood resulting in an approximate
two-fold increase directly prior to the 10th exposure (p = 0.02).
CD45
dim
/CD34
+
/CD133
SPCs continued to increase during the 3 days
following the end of exposures and increased to three-fold, 72 h aer
the 10th exposure (p = 0.008); (Figure2A).
Decreased frequency of CD45dim/CD34/CD133+
after hyperbaric exposure
While CD45dim/CD34+/CD133 SPCs increased, the frequency of
CD45
dim
/CD34
/CD133
+
primitive pro-angiogenic stem progenitor
cells signicantly decreased aer exposure to intermittent hyperbaric
air. CD45dim/CD34/CD133+ decreased by nearly ve-fold prior to the
10th exposure (p = 0.02) and mobilization decreased by six-fold 72 h
aer the 10th exposure (p = 0.01) (Figure2B).
FIGURE1
Experimental protocol.
FIGURE2
Frequency of CD45dim/CD34+/CD133-, CD45dim/CD34-/CD133+ and CD45dim/CD31+/CD105-after intermittent hyperbaric air exposure detected by flow
cytometry. (A) CD34+ and CD133- stem progenitor cells. (B) CD34- and CD133+ stem progenitor cells. (C) CD31+ and CD105- stem progenitor cells.
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 04 frontiersin.org
FIGURE3
Frequency of CD45+/CD34+/CD133-, CD45+/CD34-/CD133+, CD45+/CD31+/CD105-, and macrophage derived chemokine after intermittent hyperbaric
air exposure detected by flow cytometry and ELISA. (A) CD34+ and CD133- stem progenitor cells. (B) CD34- and CD133+ stem progenitor cells.
(C) CD31+ and CD105- stem progenitor cells. (D) Macrophage derived chemokine.
Increased frequency of CD45dim/CD31+/CD105
after hyperbaric exposure
e frequency of CD45dim/CD31
+
/CD105
-
was also
signicantly increased aer hyperbaric exposure by nearly two-fold
prior to the 10th exposure (p = 0.023) and increased to over
two-fold 3 days aer the 10th exposure (p = 0.016) (Figure2C).
Mobilization of CD45+/CD34+/CD133
The expression of CD45
+
/CD34
+
/CD133
-
also increased by
nearly 3.5 fold after 9 full exposures (p = 0.012) and increased to
over four fold 72 h following the end of the 10th exposure
(p = 0.002) (Figure3A).
Decreased frequency of CD45+/CD34/
CD133+
A signicant reduction in the expression of CD45
+
/CD34
/
CD133+ was noted resulting in a six-fold reduction prior to the 10th
exposure (p = 0.016) and recovering to 5.4-fold 72 h aer the 10th and
nal exposure (p = 0.016) (Figure3B).
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 05 frontiersin.org
Mobilization of CD45+/CD31+/CD105
e expression of CD45
+
/CD31
+
/105
-
increased by nearly two-fold
aer 9 full exposures (p = 0.02) and remained at the level through 72 h
following the end of the 10th exposure (p = 0.008) (Figure3C).
Complete results from flow cytometry
Complete results from ow cytometry testing, including
non-signicant ndings are included in tabular form for reference.
(Table2).
Macrophage-derived chemokine (MDC)
expression significantly decreased
Results from the Invitrogen ProcartaPlex Human Immune
Monitoring Panel 65-Plex ELISA like analysis showed there was only
one change to report. e expression of Macrophage-derived
Chemokine (MDC) was signicantly lower between the second and
third time points (p = 0.008). All other tests revealed no change
(Figure3D).
Complete blood count with dierential
ere were no signicant changes in CBC at any time points
(Table2).
Discussion
e smallest dose of hyperbaric air that will result in a therapeutic
eect is unknown and strongly debated among scientists and physicians.
Pressures below 1.4 atmospheres absolute of hyperbaric air are accepted
as a placebo but have not previously been tested in humans. Studies using
hyperbaric air as a placebo have resulted in a “placebo” or “participation
eect (79). is placebo eect nding is vigorously disputed because
hyperbaric air signicantly increases the partial pressure of oxygen (and
nitrogen) in the inspired air (2227). e “placebo eect” ndings have
eectively restricted the use of HBOT for many conditions including
Traumatic Brain Injury in soldiers returning from combat.
In this study weasked the question, will a small dose of hyperbaric
air (1.27 ATA), that is below the accepted 1.4 ATA therapeutic
threshold, mobilize stem cells similar to HBOT? Wehypothesized that
stem cells would bemobilized.
Indeed, stem cells were signicantly mobilized, refuting previous
“placebo eect” ndings. Weintend that the results will provide
needed experimental data to medical societies and journal editors,
and in turn provide guidance to FDA and physicians.
Critical analysis of major findings
Testing for stem cell mobilization using ow cytometry and gated by
blinded scientists, the major nding of this study is that 1.27 ATA
hyperbaric air mobilizes CD34+/CD133- SPCs and CD31+/CD105- stem
cells in humans receiving 10 daily 90-min exposures (Figures 2A,C,
respectively). Mobilization of CD34
+
SPCs has also been observed in
isobaric hyperoxic (21) and in hyperbaric hyperoxic conditions (28, 29).
e mechanism of hyperbaric air SPC mobilization in this
experiment is beyond the scope of this study, but may besimilar to
SPC mobilization found in hyperbaric oxygen therapy, which activates
nitric oxide synthase and plays a prime role in initiating CD34
+
SPC
mobilization (3033). (CD34 background) CD34
+
adult stem/
progenitor cells are a group of specic cell types that possess the
abilities of self-renewal and multipotent dierentiation (34, 35).
CD34
+
is expressed on hematopoietic and pro-angiogenic stem
progenitor cells and on endothelium (36). Pro-angiogenic stem/
progenitor cells contribute to neovascularization by a process of
homing to ischemic tissue called vasculogenesis, and by budding
endothelium from established blood vessels in a process called
angiogenesis (37, 38). Further research is needed to understand the
homing and function of the CD34
+
SPCs mobilized by intermittent
hyperbaric air exposures.
A second signicant nding of this study, which wereport for the
rst time, is hyperbaric air mobilizes CD31
+
/CD105
-
SPCs. is novel
nding has many implications in the eld of hyperbaric air and
hyperbaric oxygen. (CD31
+
background) CD31 is thought to have a
protective role in experimental atherosclerosis (39). e therapeutic
potential of CD31 agonists to manage atherosclerotic disease
manifestations is a consistent nding in pre-clinical studies (40).
Although this is the rst time that CD31 has been reported to
bemobilized by either hyperbaric air or hyperbaric oxygen, it is not
unexpected, given that CD31 is associated with endothelial function
much like CD34 and CD133. It was beyond the scope of this research
project to determine if these mobilized CD31 stem cells participate in
wound healing and angiogenesis. Future research testing the eect of
hyperbaric air mobilized CD31 stem cells on healing, angiogenesis,
and atherosclerotic disease states would beprudent.
Our results also revealed an interesting relationship between SPCs
expressing CD45
dim
/CD34
+
/CD133
and those SPCs expressing CD45
dim
/
TABLE2 Summary of cell type frequency after initial and intermittent
exposure detected by flow cytometry.
CD45dim CD45+CD45
CD45dim =,=,=,=
CD45+=,=,=,=
CD45_=,=,,=
CD31_ CD105+=,=,=,= =,=,=,= =,=,=,=
CD31+ CD105_=,=,,=,=,,=,=,=,=
CD31+ CD105+=,=,=,= =,=,=,= =,=,=,=
CD31_ CD105_=,=,=,= =,=,=,= =,=,=,=
CD34_ CD133+=,=,,=,=,,=,=,=,=
CD34+ CD133_=,=,,=,=,,=,=,=,=
CD34+ CD133+=,=,=,= =,=,=,= =,=,=,=
CD34_ CD133_=,=,=,= =,=,=,= =,=,=,=
Results from four time points represented by change symbol separated by commas.
e = symbol represents no change in frequency, the symbol represents a signicant
reduction in frequency, and the symbol indicates a signicant mobilization. e rst time
point is the control point taken prior to exposure. e second is taken directly aer the rst
exposure. e third timepoint was taken just prior to the 10th and nal exposure and the
fourth time point is the frequency 3 days aer cessation of exposures.
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 06 frontiersin.org
CD34
/CD133
+
. CD133+ SPCs are hematopoietic precursors to CD34
+
and almost all hematopoietic pluripotent and committed stem cells in
colony-forming assays express CD34+ (36). In this study wefound that
CD34+ SPCs increased while at the same time CD133+ SPCs decreased.
Wehypothesize that the exposure to intermittent hyperbaric air may
mobilize CD133
+
from bone marrow, and also play a role in the
dierentiation of the CD133+ primitive hematopoietic precursor into the
CD34
+
SPC. However, it has also been shown that CD133
+
have a high
capacity to dierentiate into other cell types including broblasts,
hepatocytes, and neural cell-types (35). Unfolding the explanation for the
signicant reduction in the expression of CD45
dim
/CD34
/CD133
+
expressing SPCs holds exciting potential.
Previous research has shown that breathing 100% oxygen at 2.4
atmospheres absolute mobilized stem/progenitor cells at a signicantly
greater rate than 2.0 Atmospheres Absolute suggesting a dose eect (41).
HBOT at 2.0 ATA (P
I
O
2
of 1426 mmHg and P
I
N
2
of 0 mmHg) resulted in
a two-fold mobilization of CD34
+
SPCs aer 1 exposure and an eight-fold
increase aer 20 exposures (28). In this study using 1.27 ATA of
hyperbaric air (P
I
O
2
of 189 mmHg and P
I
N
2
of 706 mmHg) we saw a
two-fold increase in SPC mobilization just prior to the 9th exposure. is
supports the hypothetical relationship between oxygen dose and stem
cell mobilization.
Another aim of this study was to determine if the stem progenitor
cell mobilization was durable. Because the stem progenitor cell
mobilization increased another fold following the end of exposures a
durable eect is likely.
Interestingly, wealso found changes in cells expressing CD45
+
cell
subtypes. ese changes were remarkably similar to changes in the
CD45dim population. CD45+ is expressed on all hematopoietic cells,
including HSCs and osteoclasts, which are of hematopoietic origin
(42), and is known as a pan-leukocyte marker.
Macrophage-derived chemokine (MDC) expressed in venous blood
signicantly decreased prior to the ninth exposure and returned to
pre-exposure levels 72 h following the nal exposure. e expression of
MDC is increased in idiopathic pulmonary brosis (43) and elevated
following resuscitation of patients aer hemorrhage. Macrophage-derived
chemokine may provide a therapeutic strategy to mitigate this
inammatory response (44). MDC is also thought to serve as a marker of
pharmacological therapy response in Major Depressive Disorder (45).
Changes in barometric and hydrostatic pressure may be a
mechanism of hyperbaric therapy. Small changes in atmospheric
pressure elicit responses in many organisms (46). However the
mechanism(s) is poorly understood. One possibility is that pressure is
sensed through hydrostatic compression of heterogeneous structures.
For instance, cells in suspension, including platelets (47, 48) and cartilage
cells (49, 50), respond to small changes in pressure ostensibly through
this mechanism. Another possibility is that cellular structures may
produce shear and strain through dierential compression. Microtubules,
actin and other cytoskeletal proteins respond to this type of local
mechanical stresses (51, 52). It should also benoted that certain animals
appear to respond to changes in atmospheric (air) pressure. For example,
pigeons exhibit changes in heart rate in response to changes of about
1 mbar, which is a typical daily uctuation in barometric pressure. But in
such cases, it is unclear how barometric pressure changes are sensed (53).
It is probable that changes in pressure have eects on human physiology.
Another possible mechanism ultimately resulting in stem cell
mobilization in this experiment is a progressive accumulation of
endogenous anti-oxidants at the cellular level. ese antioxidants accrue
in response to repeated exposure to a hyperoxic environment. e result
is an up-regulation of the Hypoxia-inducible factor 1α (HIF-1α)
transcription factor activity (5460). is hypothetical mechanism is
best described as the Normoxic Hypoxic Paradox. is mechanism is
best characterized by an increase in reactive oxygen species (ROS) due
to the hyperoxic cellular environment. e increased cellular ROS
creates an imbalance of ROS/scavenger antioxidants ratio. e increased
ROS molecules initially hydroxylate most of the HIF-1α, facilitating
ubiquitination and degradation of most of the HIF-1α subunits.
However, it is postulated that an adaptive response to repeated
hyperoxia, increases the production of scavengers in proportion to the
increased ROS generation. e ROS/scavenger ratio gradually becomes
balanced in the hyperoxic environment. However, when the hyperoxic
exposure ends and the cell returns to a normoxic state, the ROS/
scavenger ratio is again imbalanced, but this time with more scavengers
than ROS, which produces a reduced ROS environment. Less ROS leads
to an increase in HIF-1α, initiating HIF transcription factor activity,
resembling a hypoxic state, but in a normoxic environment. In this
study CD34+/CD133- expression trended downward directly following
the initial hyperbaric air exposure, but increased signicantly prior to
the tenth exposure. e opposite was true with CD34
-
/CD133
+
. ese
results support the hypothetical Normoxic Hypoxic Paradox mechanism.
While our study clearly showed that the hyperbaric air dose that
was used as a placebo mobilizes proangiogenic and hematopoietic
stem progenitor cells and likely has a therapeutic physiologic eect
similar to hyperbaric oxygen therapy (29, 6163), it was beyond the
scope of this study to determine clinical signicance. Hopefully our
results will generate renewed interest in hyperbaric air and future
studies will investigate both mechanism and clinical outcomes.
Limitations
e major limitation of our study is a relatively small sample size.
Future directions
It should not beoverlooked that although weincreased the
oxygen partial pressure and presume that oxygen is the active
element in hematopoietic and pro-angiogenic stem progenitor
mobilization, the partial pressure of nitrogen and remaining trace
gasses are also increased. Nitrogen is not an inert element and
can form five potential oxidation states and three potential
reduction states with various levels of reactivity (64). Recent
evidence suggests that intracellular reactive oxygen and nitrogen
species play an important role in intracellular signaling cascades
(65). However, little is known about the effect of nitrogen and
trace gasses on SPC mobilization or cytokine, chemokine and
growth factor modulation. Increased nitrogen and trace gasses as
a stimulator of SPC mobilization and chemokine modulation is a
possible mechanism in this research project and an exciting idea
to explore in future research.
It is very possible that stem cell mobilization by hyperbaric air
provides an additional modality to heal injuries. Because of its reduced
cost of delivery, it may prove benecial in developing nations and in
underserved populations. Hyperbaric air also increases safety because
the oxygen partial pressure is relatively low. e low oxygen partial
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 07 frontiersin.org
pressure in hyperbaric air may also provide increased therapeutic value
by not overloading the cerebral energy metabolism balance (66, 67).
Finally, our data suggests that the therapeutic dose of oxygen
begins at a much smaller partial pressure than previously thought and
adds a new data point on the initial portion of the hormetic curve of
oxygen dose.
Although wefound that intermittent small increases in hyperbaric
air pressure mobilized stem cells, this study should not betaken as an
endorsement of the use of intermittent hyperbaric air for any purpose
other than the indications approved by the FDA.
Why are these ndings important? First, prior to this research it was
not known that breathing a small increase in hyperbaric air would
mobilize stem progenitor cells. is knowledge could bean important
low-cost healthcare option when hyperbaric oxygen is not available,
especially in underserved populations, remote areas, and developing
nations. Second, because the technology is lightweight and portable, it is
hypothetically possible to beused when transporting combatants and
civilians out of a warzone to extend the viability of damaged tissue and to
reduce exacerbating gas embolism when air-liing by high altitude ights
is required. Finally, this research refutes the ndings of a placebo eect in
the decades-old use of slightly pressurized room air as a placebo in
hyperbaric oxygen research.
Conclusion and impact
In this study wedemonstrate for the rst time that intermittent
exposure to ostensibly insignicant pressures of hyperbaric air
mobilizes stem progenitor cells in a similar manner to that seen in
isobaric hyperoxia and hyperbaric oxygen therapy (21, 28, 29, 41).
Wealso establish that the stem progenitor cell mobilization is durable.
This research reveals that hyperbaric air, even at an ostensibly
insignificant dose, has significant effects on human physiology,
is not a placebo, and should beconsidered as an active physiologic
intervention. Its use as a pharmaceutical should
beinvestigated further.
Although this study did not test for clinical results, clinical
outcomes of hyperbaric air have been reported in similar
hyperbaric air studies (68). This study supports the data, but
refutes the conclusions in those studies by revealing a mechanism
of action for the clinical improvements reported in the hyperbaric
air group in those studies. Much more work is needed to develop
protocols of hyperbaric air dose that provide the maximum
therapeutic benefit.
ese ndings substantiate the need for testing hyperbaric air doses
prior to using hyperbaric air as a placebo in scientic investigations.
ese ndings also substantiate the urgent need for reevaluation of
ndings in historical studies using hyperbaric air placebos. Our ndings
suggest that these historical placebo-controlled studies were not placebo-
controlled studies. Paradoxically, our ndings indicate that they were
dose studies and the ndings of a “placebo eect” or “participation eect”
are inherently awed. e “ndings” and “conclusions” in studies using
hyperbaric air as a placebo should bereevaluated from a dose study
perspective. Finally, wehope the ndings in this study will persuade the
medical societies around the world to consider reevaluation of their
denition of hyperbaric medicine to include nominal hyperbaric air.
Looking back at the “Hyperbaric Air” work of Henshaw, Simpson
and Cunningham, our ndings support their reports.
Data availability statement
e original contributions presented in the study are included in
the article/supplementary material, further inquiries can bedirected
to the corresponding author.
Ethics statement
e studies involving human participants were reviewed and
approved by the University of Wisconsin–Madison. e patients/
participants provided their written informed consent to participate in
this study.
Author contributions
KM and GB: ideas conception. KM, GB, RB, and ME: study and
experiments design. KM, JM, RB, JL, and MM: experiments perform,
data acquisition, and analysis. GB and RB: guidance and critical
feedback on data acquisition and analysis. RB and ME: project
supervision. KM: manuscript writing. All authors provided critical
feedback and contributed to the nal version of manuscript.
Funding
is work was supported by the Foundation for the Study of
Inammatory Disease and the International Hyperbaric Association.
Acknowledgments
e authors would like to thank the study participants for their time
and dedication to complete this study. anks also goes to Caleb and
Sidney Gates, Dr. Rob Beckman, Dr. Vivek Balasubramanium, Dr. Barb
Bendlin, Dr. Robert Stone, Dr. William Schrage, Dr. Paul Harch, Dr.
Phillip James, Dr. Don Wollheim, Dr. Donata Oertel, Dr. Awni Al-Subu,
Dr Kara Goss, Dr. Arij Beshish, Dr. Andrew Watson, Dr. Xavier Figueroa,
Dr. Daphne Denham, Tom Fox, Freya Christensen, the University of
Wisconsin CCC Flow Cytometry Lab, Dagna Sherrar, Lauren
Nettenstrom, Kathryn Fox and Zach Sterneson, the Univeristy of
Wisconsin-Madison Department of Pediatrics and Gradaute School, Dr.
Ellen Wald, Dr. James Gern, and Kim Stevenson for the support.
Conflict of interest
e authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could
beconstrued as a potential conict of interest.
Publisher’s note
All claims expressed in this article are solely those of the
authors and do not necessarily represent those of their affiliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 08 frontiersin.org
References
1. Jain K. Textbook of hyperbaric medicine. Göttingen: Hogrefe Verlag GmbH & Co.
KG (2004).
2. Simpson A. Compressed Air as a erapeutic Agent in the Treatment of Consumption,
Asthma, Chronic Bronchitis, and Other Diseases. Glasgow Med J. (1857) 5:94–6.
3. Sellers L.M. e fallibility of the FORRESTIAN principle. "semper primus PERVENIO
maxima cum Vi". (ORVAL JAMES Cunningham). Transactions of the American
Laryngological, Rhinological and Otological Society, Inc 23, 385–405 (1964).
4. Harch P. New scientic denitions: hyperbaric therapy and hyperbaric oxygen
therapy. Med Gas Res. (2023) 13:92–3. doi: 10.4103/2045-9912.356475
5. Trimble VH. e uncertain miracle: hyperbaric oxygenation. Garden City, NY:
Doubleday & Company, Inc (1974).
6. Collet JP, Vanasse M, Marois P, Amar M, Goldberg J, Lambert J, et al. Hyperbaric
oxygen for children with cerebral palsy: a randomised multicentre trial. HBO-CP
research group. Lancet. (2001) 357:582–6. doi: 10.1016/S0140-6736(00)04054-X
7. Miller RS, Weaver LK, Bahraini N, Churchill S, Price RC, Skiba V, et al. Eects of
hyperbaric oxygen on symptoms and quality of life among service members with
persistent postconcussion symptoms: a randomized clinical trial. JAMA Intern Med.
(2015) 175:43–52. doi: 10.1001/jamainternmed.2014.5479
8. Wolf G, Cifu D, Baugh L, Carne W, Profenna L. e eect of hyperbaric oxygen on
symptoms aer mild traumatic brain injury. J Neurotrauma. (2012) 29:2606–12. doi:
10.1089/neu.2012.2549
9. Cifu DX, Walker WC, West SL, Hart BB, Franke LM, Sima A, et al. Hyperbaric
oxygen for blast-related postconcussion syndrome: three-month outcomes. Ann Neurol.
(2014) 75:277–86. doi: 10.1002/ana.24067
10. Harch PG, Fogarty EF. Subacute normobaric oxygen and hyperbaric oxygen
therapy in drowning, reversal of brain volume loss: a case report. Med Gas Res. (2017)
7:144–9. doi: 10.4103/2045-9912.208521
11. Harch PG, Fogarty EF, Staab PK, Van Meter K. Low pressure hyperbaric oxygen
therapy and SPECT brain imaging in the treatment of blast-induced chronic traumatic
brain injury (post-concussion syndrome) and post traumatic stress disorder: a case
report. Cases J. (2009) 2:6538. doi: 10.1186/1757-1626-0002-0000006538
12. Harch PG, Andrews SR, Fogarty EF, Amen D, Pezzullo JC, Lucarini J, et al. A phase
I study of low-pressure hyperbaric oxygen therapy for blast-induced post-concussion
syndrome and post-traumatic stress disorder. J Neurotrauma. (2012) 29:168–85. doi: 10.1089/
neu.2011.1895
13. Hadanny A, Catalogna M, Yaniv S, Stolar O, Rothstein L, Shabi A, et al. Hyperbaric
oxygen therapy in children with post-concussion syndrome improves cognitive and
behavioral function: a randomized controlled trial. Sci Rep. (2022) 12:15233. doi:
10.1038/s41598-022-19395-y
14. Hadanny A, Rittblat M, Bitterman M, May-Raz I, Suzin G, Boussi-Gross R,
et al. Hyperbaric oxygen therapy improves neurocognitive functions of post-stroke
patients- a retrospective analysis. Restor Neurol Neurosci. (2020) 38:93–107. doi:
10.3233/RNN-190959
15. Doenyas-Barak K, Catalogna M, Kutz I, Levi G, Hadanny A, Tal S, et al. Hyperbaric
oxygen therapy improves symptoms, brain's microstructure and functionality in veterans
with treatment resistant post-traumatic stress disorder: a prospective, randomized,
controlled trial. PLoS One. (2022) 17:e0264161. doi: 10.1371/journal.pone.0264161
16. Hadanny A, Golan H, Fishlev G, Bechor Y, Volkov O, Suzin G, et al. Hyperbaric oxygen
can induce neuroplasticity and improve cognitive functions of patients suering from anoxic
brain damage. Restor Neurol Neurosci. (2015) 33:471–86. doi: 10.3233/RNN-150517
17. Boussi-Gross R, Golan H, Volkov O, Bechor Y, Hooen D, Schnaider Beeri M,
et al. Improvement of memory impairments in poststroke patients by hyperbaric oxygen
therapy. Neuropsychology. (2015) 29:610–21. doi: 10.1037/neu0000149
18. Tal S, Hadanny A, Sasson E, Suzin G, Efrati S. Hyperbaric oxygen therapy can
induce angiogenesis and regeneration of nerve bers in traumatic brain injury patients.
Front Hum Neurosci. (2017) 11:508. doi: 10.3389/fnhum.2017.00508
19. Boussi-Gross R, Golan H, Fishlev G, Bechor Y, Volkov O, Bergan J, et al.
Hyperbaric oxygen therapy can improve post concussion syndrome years aer mild
traumatic brain injury- randomized prospective trial. PLoS One. (2013) 8:e79995. doi:
10.1371/journal.pone.0079995
20. Efrati S, Fishlev G, Bechor Y, Volkov O, Bergan J, Kliakhandler K, et al. Hyperbaric
oxygen induces late neuroplasticity in post stroke patients--randomized, prospective
trial. PLoS One. (2013) 8:e53716. doi: 10.1371/journal.pone.0053716
21. MacLaughlin K, Barton G, Braun R, Eldridge M. Eect of intermittent hyperoxia
on stem cell mobilization and cytokine expression. Med Gas Res. (2019) 9:139–44. doi:
10.4103/2045-9912.266989
22. Harch PG. Systematic review and dosage analysis: hyperbaric oxygen therapy
ecacy in mild traumatic brain injury persistent Postconcussion syndrome. Front
Neurol. (2022) 13:815056. doi: 10.3389/fneur.2022.815056
23. Hoge CW, Jonas WB. e ritual of hyperbaric oxygen and lessons for the treatment
of persistent postconcussion symptoms in military personnel. JAMA Intern Med. (2015)
175:53–4. doi: 10.1001/jamainternmed.2014.3375
24. Hoge CW, Jonas WB. Hyperbaric oxygen treatment for persistent Postconcussion
symptoms--reply. JAMA Intern Med. (2015) 175:1241. doi: 10.1001/
jamainternmed.2015.1051
25. Hu Q, Manaenko A, Guo Z, Huang L, Tang J, Zhang JH. Hyperbaric oxygen
therapy for post concussion symptoms: issues may aect the results. Med Gas Res. (2015)
5:10. doi: 10.1186/s13618-015-0033-3
26. Marois P, Mukherjee A, Ballaz L. Hyperbaric oxygen treatment for persistent
Postconcussion symptoms--a placebo eect? JAMA Intern Med. (2015) 175:1239–40.
doi: 10.1001/jamainternmed.2015.1032
27. Miller RS, Weaver LK, Brenner LA. Hyperbaric oxygen treatment for persistent
Postconcussion symptoms--reply. JAMA Intern Med. (2015) 175:1240–1. doi: 10.1001/
jamainternmed.2015.1048
28. om SR, Bhopale VM, Velazquez OC, Goldstein LJ, om LH, Buerk DG. Stem
cell mobilization by hyperbaric oxygen. Am J Physiol Heart Circ Physiol. (2006)
290:H1378–86. doi: 10.1152/ajpheart.00888.2005
29. om SR, Milovanova TN, Yang M, Bhopale VM, Sorokina EM, Uzun G, 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) 19:149–61. doi:
10.1111/j.1524-475X.2010.00660.x
30. Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau-Ihling K,
et al. Essential role of endothelial nitric oxide synthase for mobilization of stem and
progenitor cells. Nat Med. (2003) 9:1370–6. doi: 10.1038/nm948
31. om SR, Fisher D, Zhang J, Bhopale VM, Ohnishi ST, Kotake Y, et al. Stimulation
of perivascular nitric oxide synthesis by oxygen. Am J Physiol Heart Circ Physiol. (2003)
284:H1230–9. doi: 10.1152/ajpheart.01043.2002
32. Thom SR, Bhopale V, Fisher D, Manevich Y, Huang PL, Buerk DG. Stimulation
of nitric oxide synthase in cerebral cortex due to elevated partial pressures of
oxygen: an oxidative stress response. J Neurobiol. (2002) 51:85–100. doi: 10.1002/
neu.10044
33. Goldstein LJ, Gallagher KA, Bauer SM, Bauer RJ, Baireddy V, Liu ZJ, et al.
Endothelial progenitor cell release into circulation is triggered by hyperoxia-induced
increases in bone marrow nitric oxide. Stem Cells. (2006) 24:2309–18. doi: 10.1634/
stemcells.2006-0010
34. Kitamura S, Yamasaki Y, Kinomura M, Sugaya T, Sugiyama H, Maeshima Y,
et al. Establishment and characterization of renal progenitor like cells from S3
segment of nephron in rat adult kidney. FASEB J. (2005) 19:1789–97. doi: 10.1096/
fj.05-3942com
35. Bachelier K, Bergholz C, Friedrich EB. Dierentiation potential and functional
properties of a CD34-CD133+ subpopulation of endothelial progenitor cells. Mol Med
Rep. (2020) 21:501–7. doi: 10.3892/mmr.2019.10831
36. Faramarz NP, Sophie XS, Ryan TP. Chapter 2 – principles of immunophenotyping.
Atlas Hematopathol. (2018):29–56. doi: 10.1016/B978-0-12-809843-1.00002-4
37. Rai S, Lyden D. erapeutic stem and progenitor cell transplantation for organ
vascularization and regeneration. Nat Med. (2003) 9:702–12. doi: 10.1038/nm0603-702
38. Pierro M, Ciarmoli E, ébaud B. Stem cell therapy for neonatal lung diseases In:
G Steinho, editor. Regenerative medicine- from protocol to patient: 4. Regenerative
therapies I. Cham: Springer International Publishing (2016). 319–57.
39. Goel R, Schrank BR, Arora S, Boylan B, Fleming B, Miura H, et al. Site-specic
eects of PECAM-1 on atherosclerosis in LDL receptor-decient mice. Arterioscler
romb Vasc Biol. (2008) 28:1996–2002. doi: 10.1161/ATVBAHA.108.172270
40. Caligiuri G. CD31 as a therapeutic target in atherosclerosis. Circ Res. (2020)
126:1178–89. doi: 10.1161/CIRCRESAHA.120.315935
41. Heyboer M 3rd, Milovanova TN, Wojcik S, Grant W, Chin M, Hardy KR, et al.
CD34+/CD45-dim stem cell mobilization by hyperbaric oxygen – changes with oxygen
dosage. Stem Cell Res. (2014) 12:638–45. doi: 10.1016/j.scr.2014.02.005
42. Shivtiel S, Kollet O, Lapid K, Schajnovitz A, Goichberg P, Kalinkovich A, et al.
CD45 regulates retention, motility, and numbers of hematopoietic progenitors, and
aects osteoclast remodeling of metaphyseal trabecules. J Exp Med. (2008) 205:2381–95.
doi: 10.1084/jem.20080072
43. Yogo Y, Fujishima S, Inoue T, Saito F, Shiomi T, Yamaguchi K, et al. Macrophage
derived chemokine (CCL22), thymus and activation-regulated chemokine (CCL17), and
CCR4 in idiopathic pulmonary brosis. Respir Res. (2009) 10:80. doi:
10.1186/1465-9921-10-80
44. Richter JR, Sutton JM, Belizaire RM, Friend LA, Schuster RM, Johannigman TA,
et al. Macrophage-derived chemokine (CCL22) is a novel mediator of lung inammation
following hemorrhage and resuscitation. Shock. (2014) 42:525–31. doi: 10.1097/
SHK.0000000000000253
45. Milenkovic VM, Sarubin N, Hilbert S, Baghai TC, Stöer F, Lima-Ojeda JM, et al.
Macrophage-derived chemokine: a putative marker of pharmacological therapy
response in major depression? Neuroimmunomodulation. (2017) 24:106–12. doi:
10.1159/000479739
MacLaughlin et al. 10.3389/fneur.2023.1192793
Frontiers in Neurology 09 frontiersin.org
46. Macdonald AG, Fraser PJ. e transduction of very small hydrostatic pressures. Comp
Biochem Physiol A Mol Integr Physiol. (1999) 122:13–36. doi: 10.1016/S1095-6433(98)10173-3
47. Torsellini A, Becucci A, Citi S, Cozzolino F, Guidi G, Lombardi V, et al. Eects of
pressure excursions on human platelets. In vitro studies on betathromboglobulin (beta-
TG) and platelet factor 4 (PF4) release and on platelet sensitivity to ADP-aggregation.
Haematologica. (1982) 67:860–6.
48. Torsellini A, Doni L, Palinski W, Guidi G, Lombardi V. Pressure-induced metabolic
activation of platelets demonstrated in vitro by determination of malondialdehyde
formation. romb Haemost. (1984) 52:4–6. doi: 10.1055/s-0038-1661123
49. Bourret LA, Rodan GA. e role of calcium in the inhibition of cAMP
accumulation in epiphyseal cartilage cells exposed to physiological pressure. J Cell
Physiol. (1976) 88:353–61. doi: 10.1002/jcp.1040880311
50. Burger EH, Klein-Nulend J, Veldhuijzen JP. Mechanical stress and osteogenesis
invitro. J Bone Miner Res. (1992) 7:S397–401. doi: 10.1002/jbmr.5650071406
51. Buxbaum RE, Heidemann SR. A thermodynamic model for force integration and
microtubule assembly during axonal elongation. J eor Biol. (1988) 134:379–90. doi:
10.1016/S0022-5193(88)80068-7
52. Wang N, Butler JP, Ingber DE. Mechanotransduction across the cell surface and
through the cytoskeleton. Science. (1993) 260:1124–7. doi: 10.1126/science.7684161
53. Kreithen ML, Keeton WT. Detection of changes in atmospheric pressure by the homing
pigeon, Columba livia. J Compar. Physiol. (1974) 89:73–82. doi: 10.1007/BF00696164
54. Balestra C, Germonpre P. Hypoxia, a multifaceted phenomenon: the example of the
"normobaric oxygen paradox". Eur J Appl Physiol. (2012) 112:4173–5. doi: 10.1007/
s00421-012-2392-y
55. Balestra C, Germonpre P, Poortmans JR, Marroni A. Serum erythropoietin levels in healthy
humans aer a short period of normobaric and hyperbaric oxygen breathing: the “normobaric
oxygen paradox”. J Appl Physiol. (2006) 100:512–8. doi: 10.1152/japplphysiol.00964.2005
56. de Bels D, eunissen S, Devriendt J, Germonpré P, Lafere P, Valsamis J, et al. e
‘normobaric oxygen paradox’: does it increase haemoglobin? Diving Hyperb Med. (2012)
42:67–71.
57. Fratantonio D, Virgili F, Zucchi A, Lambrechts K, Latronico T, Lafère P, et al. Increasing
oxygen partial pressures induce a distinct transcriptional response in human PBMC: a pilot
study on the “normobaric oxygen paradox”. Int J Mol Sci. (2021) 22:458. doi: 10.3390/
ijms22010458
58. Hadanny A, Efrati S. e hyperoxic-hypoxic paradox. Biomol er. (2020) 10:958.
doi: 10.3390/biom10060958
59. Lafere P, Schubert T, De Bels D, Germonpre P, Balestra C. Can the normobaric
oxygen paradox (NOP) increase reticulocyte count aer traumatic hip surgery? J Clin
Anesth. (2013) 25:129–34. doi: 10.1016/j.jclinane.2012.06.021
60. Rocco M, D'Itri L, De Bels D, Corazza F, Balestra C. e “normobaric oxygen
paradox”: a new tool for the anesthetist? Minerva Anestesiol. (2014) 80:366–72.
61. Fosen KM, om SR. Hyperbaric oxygen, vasculogenic stem cells, and wound
healing. Antioxid Redox Signal. (2014) 21:1634–47. doi: 10.1089/ars.2014.5940
62. om SR, Hampton M, Troiano MA, Mirza Z, Malay DS, Shannon S, et al.
Measurements of CD34+/CD45-dim stem cells predict healing of diabetic neuropathic
wounds. Diabetes. (2016) 65:486–97. doi: 10.2337/db15-0517
63. Gallagher KA, Goldstein LJ, om SR, Velazquez OC. Hyperbaric oxygen and
bone marrow-derived endothelial progenitor cells in diabetic wound healing. Vascular.
(2006) 14:328–37. doi: 10.2310/6670.2006.00057
64. Marozkina NV, Gaston B. Nitrogen chemistry and lung physiology. Annu Rev
Physiol. (2015) 77:431–52. doi: 10.1146/annurev-physiol-021113-170352
65. Weidinger A, Kozlov AV. Biological activities of reactive oxygen and nitrogen
species: oxidative stress versus signal transduction. Biomol er. (2015) 5:472–84. doi:
10.3390/biom5020472
66. Holbach KH, Schröder FK, Köster S. Alterations of cerebral metabolism in cases
with acute brain injuries during spontaneous respiration of air, oxygen and hyperbaric
oxygen. Eur Neurol. (1972) 8:158–60. doi: 10.1159/000114570
67. Holbach KH, Caroli A, Wassmann H. Cerebral energy metabolism in patients with
brain lesions at normo- and hyperbaric oxygen pressures. J Neurol. (1977) 217:17–30.
doi: 10.1007/BF00316313
... Moreover, studies have shown that stem cell mobilization is significantly increased following HBT. MacLaughlin et al. (8) reported that an exposition to hyperbaric air at 1.27 ATA generates up to a three-fold increase in circulating stems cells. Finally, HBT has been shown to up-or down-regulated the expression of thousands of genes; the largest clusters of upregulated genes were the anti-inflammatory genes and those that coded for growth and repair hormone, and the largest clusters of downregulated genes were the pro-inflammatory genes and apoptotic genes (9,10). ...
... The controversy on the efficacity of HBT in CP is still going on as some studies have wrongly considered mild hyperbaric pressures as a sham treatment for control groups (11,12). These claims have been increasingly contested as many powerful healing mechanisms are activated even at very low pressure (8,(13)(14)(15). ...
... This lower level of oxygen paired with the 1.5 ATA pressurization has been attempted to replicate the levels of oxygen perfusion that would normally be observed under ambient air. However, pressurization alone induce many physiological changes regardless of oxygenation, and it has been shown repeatedly that many powerful healing mechanisms can be activated even with a limited pressure increase (5,8,14,15). For this reason, it is inaccurate to consider any group receiving HBT as a control, regardless of their levels of oxygenation (8,(71)(72)(73). ...
Article
Full-text available
The Gross Motor Function Measure is used in most studies measuring gross motor function in children with cerebral palsy. In many studies, including those evaluating the effect of hyperbaric treatment, the Gross Motor Function Measure variations were potentially misinterpreted because of the lack of control groups. The Gross Motor Function Measure Evolution Ratio (GMFMER) uses historical data from the Gross Motor Function Classification System curves and allows to re-analyze previous published studies which used the Gross Motor Function Measure by considering the natural expected evolution of the Gross Motor Function Measure. As the GMFMER is defined by the ratio between the recorded Gross Motor Function Measure score increase and the expected increase attributed to natural evolution during the duration of the study (natural evolution yields a GMFMER of 1), it becomes easy to assess and compare the efficacy of different treatments. Objective The objective of this study is to revisit studies done with different dosage of hyperbaric treatment and to compare the GMFMER measured in these studies with those assessing the effects of various recommended treatments in children with cerebral palsy. Methods PubMed Searches were conducted to included studies that used the Gross Motor Function Measure to evaluate the effect of physical therapy, selective dorsal rhizotomy, botulinum toxin injection, hippotherapy, stem cell, or hyperbaric treatment. The GMFMER were computed for each group of the included studies. Results Forty-four studies were included, counting 4 studies evaluating the effects of various dosage of hyperbaric treatment in children with cerebral palsy. Since some studies had several arms, the GMFMER has been computed for 69 groups. The average GMFMER for the groups receiving less than 2 h/week of physical therapy was 2.5 ± 1.8 whereas in context of very intensive physical therapy it increased to 10.3 ± 6.1. The GMFMER of stem cell, selective dorsal rhizotomy, hippotherapy, and botulinum toxin treatment was, 6.0 ± 5.9, 6.5 ± 2.0, 13.3 ± 0.6, and 5.0 ± 2.9, respectively. The GMFMER of the groups of children receiving hyperbaric treatment were 28.1 ± 13.0 for hyperbaric oxygen therapy and 29.8 ± 6.8 for hyperbaric air. Conclusion The analysis of the included studies with the GMFMER showed that hyperbaric treatment can result in progress of gross motor function more than other recognized treatments in children with cerebral palsy.
... Frequently, this method utilizes pressures between 2 to 3 ATA. Nevertheless, promising results have also been obtained for certain conditions with pressures less than 2 ATA (1.5 ATA) [2,3], and in some studies, even 'hyperbaric air' seems to be of interest [4]. While some protocols accept the use of 6 ATA (e.g., for the treatment of gas embolism), little benefit is usually reported from pressures above 3 ATA as this may be associated with a plethora of adverse effects [5]. ...
... The need for several sessions to reach a relevant effect is likewise commonly accepted; however, the optimal hyperbaric oxygen levels and the time needed between each session to optimize cellular responses-such as Hypoxia inducible factor (HIF-1α) or nuclear factor kappa β (NF-Kβ), erythroid related factor 2 (NRF2), cellular vesicles and microparticles, Caspase 3 [7][8][9][10]-are still debated and stand solely on observational clinical outcomes. Some recent experimental works have been evaluating the effects of different levels of oxygen on oxidative stress under hypoxic [11][12][13][14], normobaric hyperoxic [15][16][17], and hyperbaric hyperoxic [4,[6][7][8] conditions. The encouraging, but also challenging, results lead even to question if some oxygen levels formerly considered as 'HBOT sham' [18] may be of therapeutic interest [4,7]. ...
... Some recent experimental works have been evaluating the effects of different levels of oxygen on oxidative stress under hypoxic [11][12][13][14], normobaric hyperoxic [15][16][17], and hyperbaric hyperoxic [4,[6][7][8] conditions. The encouraging, but also challenging, results lead even to question if some oxygen levels formerly considered as 'HBOT sham' [18] may be of therapeutic interest [4,7]. ...
Article
Full-text available
Hyperbaric oxygen therapy (HBOT) is a therapeutical approach based on exposure to pure oxygen in an augmented atmospheric pressure. Although it has been used for years, the exact kinetics of the reactive oxygen species (ROS) between different pressures of hyperbaric oxygen exposure are still not clearly evidenced. In this study, the metabolic responses of hyperbaric hyperoxia exposures for 1 h at 1.4 and 2.5 ATA were investigated. Fourteen healthy non-smoking subjects (2 females and 12 males, age: 37.3 ± 12.7 years old (mean ± SD), height: 176.3 ± 9.9 cm, and weight: 75.8 ± 17.7 kg) volunteered for this study. Blood samples were taken before and at 30 min, 2 h, 24 h, and 48 h after a 1 h hyperbaric hyperoxic exposure. The level of oxidation was evaluated by the rate of ROS production, nitric oxide metabolites (NOx), and the levels of isoprostane. Antioxidant reactions were assessed through measuring superoxide dismutase (SOD), catalase (CAT), cysteinylglycine, and glutathione (GSH). The inflammatory response was measured using interleukine-6, neopterin, and creatinine. A short (60 min) period of mild (1.4 ATA) and high (2.5 ATA) hyperbaric hyperoxia leads to a similar significant increase in the production of ROS and antioxidant reactions. Immunomodulation and inflammatory responses, on the contrary, respond proportionally to the hyperbaric oxygen dose. Further research is warranted on the dose and the inter-dose recovery time to optimize the potential therapeutic benefits of this promising intervention.
... Frontiers in Aging frontiersin.org 05 has been found to be varied in different studies (Balestra et al., 2022;de Wolde et al., 2022;Leveque et al., 2023;MacLaughlin et al., 2023). Leveque et al. (2023), reported comparable levels of ROS, upon 30 min of HBOT under 1.4 ATA and 2.5 ATA of 100% oxygen which peaks after 2 h of treatment. ...
Article
Full-text available
Hyperbaric Oxygen Therapy (HBOT) utilizes 100% oxygen at high atmospheric pressure for clinical applications. HBOT has proven to be an effective supplementary treatment for a variety of clinical and pathological disorders. HBOT’s therapeutic results are based on the physiological effects of increased tissue oxygenation, or improved oxygen bioavailability. HBOT’s current indications in illnesses like as wound healing, thermal or radiation burns, and tissue necrosis point to its function in facilitating the regeneration process. Various research has revealed that HBOT plays a function in vascularization, angiogenesis, and collagen production augmentation. Individual regeneration capacity is influenced by both environmental and genetic factors. Furthermore, the regenerating ability of different types of tissues varies, and this ability declines with age. HBOT affects physiological processes at the genetic level by altering gene expression, delaying cell senescence, and assisting in telomere length enhancement. The positive results in a variety of indications, ranging from tissue regeneration to better cognitive function, indicate that it has enormous potential in regenerative and anti-aging therapy.
... It is known that oxygen exposure elicits "Oxy-Inflammation", a term proposed by Valacchi et al. [53] for a condition characterized by the alteration of systemic inflammation and severely compromised redox balance, and we can understand why several hyperbaric sessions (at 2.5 ATA) are needed to adapt and counteract inflammatory or oxidative stress [54]. Lower oxygen levels or even "hyperbaric air" have faster cellular hormetic responses [33,55]. ...
Article
Full-text available
The "normobaric oxygen paradox" (NOP) describes the response to the return to normoxia after a hyperoxic event, sensed by tissues as an oxygen shortage, up-regulating redox-sensitive transcription factors. We have previously characterized the time trend of oxygen-sensitive transcription factors in human PBMCs, in which the return to normoxia after 30% oxygen is sensed as a hypoxic trigger, characterized by hypoxia-induced factor (HIF-1) activation. On the contrary, 100% and 140% oxygen induce a shift toward an oxidative stress response, characterized by NRF2 and NF-kB activation in the first 24 h post exposure. Herein, we investigate whether this paradigm triggers Advanced Glycation End products (AGEs) and Advanced Oxidation Protein Products (AOPPs) as circulating biomarkers of oxidative stress. Secondly, we studied if mitochondrial biogenesis was involved to link the cellular response to oxidative stress in human PBMCs. Our results show that AGEs and AOPPs increase in a different manner according to oxygen dose. Mitochondrial levels of peroxiredoxin (PRX3) supported the cellular response to oxidative stress and increased at 24 h after mild hyperoxia, MH (30% O 2), and high hyperoxia, HH (100% O 2), while during very high hyperoxia, VHH (140% O 2), the activation was significantly high only at 3 h after oxygen exposure. Mitochondrial biogenesis was activated through nuclear translocation of PGC-1α in all the experimental conditions. However, the consequent release of nuclear Mitochondrial Transcription Factor A (TFAM) was observed only after MH exposure. Conversely, HH and VHH are associated with a progressive loss of NOP response in the ability to induce TFAM expression despite a nuclear translocation of PGC-1α also occurring in these conditions. This study confirms that pulsed high oxygen treatment elicits specific cellular responses, according to its partial pressure and time of administration, and further emphasizes the importance of targeting the use of oxygen to activate specific effects on the whole organism.
... In 2018, the VA Evidence-Based Synthesis Program for traumatic brain injury (TBI) and/or PTSD stated that based on the data available up to 2018, it was difficult to make clear decisions regarding the use of HBOT for TBI and PTSD (Peterson et al., 2018). However, since 2018 preclinical as well as clinical data accumulated (Deru et al., 2018;Lin et al., 2019;Lippert and Borlongan, 2019;Mozayeni et al., 2019;Harch et al., 2020;Doenyas-Barak et al., 2022Hadanny et al., 2022;MacLaughlin et al., 2023) and contributed to our understanding of the potential role of HBOT in PTSD treatment. ...
Article
Full-text available
Post-traumatic stress disorder (PTSD) affects up to 30% of veterans returning from the combat zone. Unfortunately, a substantial proportion of them do not remit with the current available treatments and thus continue to experience long-term social, behavioral, and occupational dysfunction. Accumulating data implies that the long-standing unremitting symptoms are related to changes in brain activity and structure, mainly disruption in the frontolimbic circuit. Hence, repair of brain structure and restoration of function could be a potential aim of effective treatment. Hyperbaric oxygen therapy (HBOT) has been effective in treating disruptions of brain structure and functions such as stroke, traumatic brain injury, and fibromyalgia even years after the acute insult. These favorable HBOT brain effects may be related to recent protocols that emphasize frequent fluctuations in oxygen concentrations, which in turn contribute to gene expression alterations and metabolic changes that induce neuronal stem cell proliferation, mitochondrial multiplication, angiogenesis, and regulation of the inflammatory cascade. Recently, clinical findings have also demonstrated the beneficial effect of HBOT on veterans with treatment-resistant PTSD. Moderation of intrusive symptoms, avoidance, mood and cognitive symptoms, and hyperarousal were correlated with improved brain function and with diffusion tensor imaging-defined structural changes. This article reviews the current data on the regenerative biological effects of HBOT, and the ongoing research of its use for veterans with PTSD.
... These encouraging but also challenging results lead us to question if some oxygen levels formerly considered as 'HBOT sham' [50,51] may be of therapeutic interest [9,51,52]. ...
Article
Full-text available
Molecular oxygen (O2) is one of the four most important elements on Earth (alongside carbon, nitrogen and hydrogen); aerobic organisms depend on it to release energy from carbon-based molecules [...]
Article
Full-text available
A 78-year-old man with dementia experienced waxing and waning of symptoms with changes in altitude as he traveled from his home in the Rocky Mountains to lower elevations and back. To replicate the improvement in his symptoms with travel to lower elevations (higher pressure), the patient was treated with a near-identical repressurization in a hyperbaric chamber using compressed air. With four 1-h treatments at 1.3 Atmospheres Absolute (ATA) and concurrent administration of low-dose oral glutathione amino acid precursors, he recovered speech and showed improvement in activities of daily living. Regional broadcast media had documented his novel recovery. Nosocomial COVID-19 and withdrawal of hyperbaric air therapy led to patient demise 7 months after initiation of treatment. It is theorized that hyperbaric air therapy stimulated mitochondrial biochemical and physical changes, which led to clinical improvement.
Book
Full-text available
Dalam buku ajar terapi oksigen hiperbarik ini, kami membawa pembaca dalam sebuah perjalanan mendalam ke dalam dunia pengobatan yang penuh harapan dan keajaiban. Seperti seorang pelaut yang mengarungi lautan luas, kami menjelajahi beragam pengetahuan dan pengalaman yang terkait dengan terapi oksigen hiperbarik. Buku ini bukan sekadar kumpulan fakta dan data, melainkan sebuah narasi yang mengalir seperti sungai yang membawa kita ke tempat-tempat yang belum pernah kita kunjungi sebelumnya. Kami berusaha menyajikan informasi secara jelas dan menggugah hati, seolah setiap kata adalah tetesan embun yang menyirami benih pengetahuan di tanah hati pembaca. Dari halaman demi halaman, pembaca akan diajak untuk memahami betapa pentingnya terapi oksigen hiperbarik dalam menyembuhkan berbagai penyakit dan kondisi medis. Seperti sinar matahari yang menerangi dunia setelah badai, terapi ini membawa harapan bagi mereka yang sedang berjuang melawan penyakit dan cobaan hidup. Kami membahas berbagai aspek terapi oksigen hiperbarik dengan mendalam dan menyeluruh, mulai dari pengaruhnya terhadap fungsi perhatian manusia hingga peranannya dalam mengobati penyakit-penyakit kronis seperti diabetes dan gangguan neurologis. Seperti seorang tabib yang cermat dan teliti, kami mengulas setiap detail dengan penuh perhatian agar pembaca dapat memahami dengan lebih baik. Namun, buku ini tidak hanya mengulas aspek-aspek ilmiah semata. Kami juga mengajak pembaca untuk merenungkan makna kehidupan dan keberanian dalam menghadapi cobaan. Seperti pepatah yang mengatakan bahwa setiap badai pasti akan berlalu, terapi oksigen hiperbarik mengajarkan kita untuk tetap bertahan dan memperjuangkan kesehatan dan kebahagiaan. Dengan kata lain, buku ini adalah sebuah cahaya di tengah kegelapan, sebuah sumber inspirasi bagi mereka yang sedang berjuang melawan penyakit dan kesulitan. Seperti bintang-bintang yang bersinar di langit malam, terapi oksigen hiperbarik memberikan harapan bagi mereka yang sedang dalam keadaan terpuruk. Dengan rendah hati, kami berharap bahwa buku ini dapat menjadi teman setia bagi para praktisi kesehatan dan pasien, serta menjadi sumber pengetahuan yang bermanfaat bagi siapa pun yang membacanya. Semoga cahaya yang kami pancarkan melalui buku ini dapat menyinari jalan bagi mereka yang membutuhkan, dan semoga kita semua dapat menjadi lebih bijaksana dan tangguh dalam menghadapi tantangan kehidupan. Terima kasih atas segala dukungan dan doa yang telah diberikan. Semoga buku ini dapat memberikan manfaat yang besar bagi kesehatan dan kesejahteraan umat manusia.
Article
Full-text available
Citation: Velikic, G.; Maric, D.M.; Maric, D.L.; Supic, G.; Puletic, M.; Dulic, O.; Vojvodic, D. Harnessing the Stem Cell Niche in Regenerative Medicine: Innovative Avenue to Combat Neurodegenerative Diseases. Int. J. Mol. Sci. 2024, 25, 993. doi: 10.3390/ijms25020993 https://www.mdpi.com/1422-0067/25/2/993 Abstract: Regenerative medicine harnesses the body's innate capacity for self-repair to restore malfunctioning tissues and organs. Stem cell therapies represent a key regenerative strategy, but to effectively harness their potential necessitates a nuanced understanding of the stem cell niche. This specialized microenvironment regulates critical stem cell behaviors including quiescence, activation, differentiation, and homing. Emerging research reveals that dysfunction within endogenous neural stem cell niches contributes to neurodegenerative pathologies and impedes regeneration. Strategies such as modifying signaling pathways, or epigenetic interventions to restore niche homeostasis and signaling, hold promise for revitalizing neurogenesis and neural repair in diseases like Alzheimer's and Parkinson's. Comparative studies of highly regenerative species provide evolutionary clues into niche-mediated renewal mechanisms. Leveraging endogenous bioelectric cues and crosstalk between gut, brain, and vascular niches further illuminates promising therapeutic opportunities. Emerging techniques like single-cell transcriptomics, organoids, microfluidics, artificial intelligence, in silico modeling, and transdifferentiation will continue to unravel niche complexity. By providing a comprehensive synthesis integrating diverse views on niche components, developmental transitions, and dynamics, this review unveils new layers of complexity integral to niche behavior and function, which unveil novel prospects to modulate niche function and provide revolutionary treatments for neurodegenerative diseases.
Article
Full-text available
Persistent post-concussion syndrome (PPCS) is a common and significant morbidity among children following traumatic brain injury (TBI) and the evidence for effective PPCS treatments remains limited. Recent studies have shown the beneficial effects of hyperbaric oxygen therapy (HBOT) in PPCS adult patients. This randomized, sham-control, double blind trial evaluated the effect of hyperbaric oxygen therapy (HBOT) on children (age 8–15) suffering from PPCS from mild-moderate TBI events six months to 10 years prior. Twenty-five children were randomized to receive 60 daily sessions of HBOT (n = 15) or sham (n = 10) treatments. Following HBOT, there was a significant increase in cognitive function including the general cognitive score (d = 0.598, p = 0.01), memory (d = 0.480, p = 0.02), executive function (d = 0.739, p = 0.003), PPCS symptoms including emotional score (p = 0.04, d = – 0.676), behavioral symptoms including hyperactivity (d = 0.244, p = 0.03), global executive composite score (d = 0.528, p = 0.001), planning/organizing score (d = 1.09, p = 0.007). Clinical outcomes correlated with significant improvements in brain MRI microstructural changes in the insula, supramarginal, lingual, inferior frontal and fusiform gyri. The study suggests that HBOT improves both cognitive and behavioral function, PPCS symptoms, and quality of life in pediatric PPCS patients at the chronic stage, even years after injury. Additional data is needed to optimize the protocol and to characterize the children who can benefit the most.
Article
Full-text available
Background Mild traumatic brain injury results in over 15% of patients progressing to Persistent Postconcussion Syndrome, a condition with significant consequences and limited treatment options. Hyperbaric oxygen therapy has been applied to Persistent Postconcussion Syndrome with conflicting results based on its historical understanding/definition as a disease-specific therapy. This is a systematic review of the evidence for hyperbaric oxygen therapy (HBOT) in Persistent Postconcussion Syndrome using a dose-analysis that is based on the scientific definition of hyperbaric oxygen therapy as a dual-component drug composed of increased barometric pressure and hyperoxia. Methods In this review, PubMed, CINAHL, and the Cochrane Systematic Review Database were searched from August 8–22, 2021 for all adult clinical studies published in English on hyperbaric oxygen therapy in mild traumatic brain injury Persistent Postconcussion Syndrome (symptoms present at least 3 months). Randomized trials and studies with symptomatic and/or cognitive outcomes were selected for final analysis. Randomized trials included those with no-treatment control groups or control groups defined by either the historical or scientific definition. Studies were analyzed according to the dose of oxygen and barometric pressure and classified as Levels 1–5 based on significant immediate post-treatment symptoms or cognitive outcomes compared to control groups. Levels of evidence classifications were made according to the Centre for Evidence-Based Medicine and a practice recommendation according to the American Society of Plastic Surgeons. Methodologic quality and bias were assessed according to the PEDro Scale. Results Eleven studies were included: six randomized trials, one case-controlled study, one case series, and three case reports. Whether analyzed by oxygen, pressure, or composite oxygen and pressure dose of hyperbaric therapy statistically significant symptomatic and cognitive improvements or cognitive improvements alone were achieved for patients treated with 40 HBOTS at 1.5 atmospheres absolute (ATA) (four randomized trials). Symptoms were also improved with 30 treatments at 1.3 ATA air (one study), positive and negative results were obtained at 1.2 ATA air (one positive and one negative study), and negative results in one study at 2.4 ATA oxygen. All studies involved <75 subjects/study. Minimal bias was present in four randomized trials and greater bias in 2. Conclusion In multiple randomized and randomized controlled studies HBOT at 1.5 ATA oxygen demonstrated statistically significant symptomatic and cognitive or cognitive improvements alone in patients with mild traumatic brain injury Persistent Postconcussion Syndrome. Positive and negative results occurred at lower and higher doses of oxygen and pressure. Increased pressure within a narrow range appears to be the more important effect than increased oxygen which is effective over a broad range. Improvements were greater when patients had comorbid Post Traumatic Stress Disorder. Despite small sample sizes, the 1.5 ATA HBOT studies meet the Centre for Evidence-Based Medicine Level 1 criteria and an American Society of Plastic Surgeons Class A Recommendation for HBOT treatment of mild traumatic brain injury persistent postconcussion syndrome.
Article
Full-text available
Introduction Post-traumatic stress disorder (PTSD) is characterized by changes in both brain activity and microstructural integrity. Cumulative evidence demonstrates that hyperbaric oxygen therapy (HBOT) induces neuroplasticity and case-series studies indicate its potentially positive effects on PTSD. The aim of the study was to evaluate HBOT’s effect in veterans with treatment resistant PTSD. Methods Veterans with treatment resistant PTSD were 1:1 randomized to HBOT or control groups. All other brain pathologies served as exclusion criteria. Outcome measures included clinician-administered PTSD scale-V (CAPS-V) questionnaires, brief symptom inventory (BSI), BECK depression inventory (BDI), brain microstructural integrity evaluated by MRI diffuse tensor imaging sequence (DTI), and brain function was evaluated by an n-back task using functional MRI (fMRI). The treatment group underwent sixty daily hyperbaric sessions. No interventions were performed in the control group. Results Thirty-five veterans were randomized to HBOT (N = 18) or control (n = 17) and 29 completed the protocol. Following HBOT, there was a significant improvement in CAPS-V scores and no change in the control (F = 30.57, P<0.0001, Net effect size = 1.64). Significant improvements were also demonstrated in BSI and BDI scores (F = 5.72, P = 0.024 Net effect size = 0.89, and F = 7.65, P = 0.01, Net effect size = 1.03). Improved brain activity was seen in fMRI in the left dorsolateral prefrontal, middle temporal gyri, both thalami, left hippocampus and left insula. The DTI showed significant increases in fractional anisotropy in the fronto-limbic white-matter, genu of the corpus callosum and fornix. Conclusions HBOT improved symptoms, brain microstructure and functionality in veterans with treatment resistant PTSD.
Article
Full-text available
The term “normobaric oxygen paradox” (NOP), describes the response to the return to normoxia after a hyperoxic event, sensed by tissues as oxygen shortage, and resulting in up-regulation of the Hypoxia-inducible factor 1α (HIF-1α) transcription factor activity. The molecular characteristics of this response have not been yet fully characterized. Herein, we report the activation time trend of oxygen-sensitive transcription factors in human peripheral blood mononuclear cells (PBMCs) obtained from healthy subjects after one hour of exposure to mild (MH), high (HH) and very high (VHH) hyperoxia, corresponding to 30%, 100%, 140% O2, respectively. Our observations confirm that MH is perceived as a hypoxic stress, characterized by the activation of HIF-1α and Nuclear factor (erythroid-derived 2)-like 2 (NRF2), but not Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-kB). Conversely, HH is associated to a progressive loss of NOP response and to an increase in oxidative stress leading to NRF2 and NF-kB activation, accompanied by the synthesis of glutathione (GSH). After VHH, HIF-1α activation is totally absent and oxidative stress response, accompanied by NF-kB activation, is prevalent. Intracellular GSH and Matrix metallopeptidase 9 (MMP-9) plasma levels parallel the transcription factors activation pattern and remain elevated throughout the observation time. In conclusion, our study confirms that, in vivo, the return to normoxia after MH is sensed as a hypoxic trigger characterized by HIF-1α activation. On the contrary, HH and VHH induce a shift toward an oxidative stress response, characterized by NRF2 and NF-kB activation in the first 24 h post exposure.
Article
Full-text available
Effective metabolism is highly dependent on a narrow therapeutic range of oxygen. Accordingly, low levels of oxygen, or hypoxia, are one of the most powerful inducers of gene expression, metabolic changes, and regenerative processes, including angiogenesis and stimulation of stem cell proliferation, migration, and differentiation. The sensing of decreased oxygen levels (hypoxia) or increased oxygen levels (hyperoxia), occurs through specialized chemoreceptor cells and metabolic changes at the cellular level, which regulate the response. Interestingly, fluctuations in the free oxygen concentration rather than the absolute level of oxygen can be interpreted at the cellular level as a lack of oxygen. Thus, repeated intermittent hyperoxia can induce many of the mediators and cellular mechanisms that are usually induced during hypoxia. This is called the hyperoxic-hypoxic paradox (HHP). This article reviews oxygen physiology, the main cellular processes triggered by hypoxia, and the cascade of events triggered by the HHP.
Article
Full-text available
The potential of CD31 as a therapeutic target in atherosclerosis has been considered ever since its cloning in the 1990s, but the exact role played by this molecule in the biologic events underlying atherosclerosis has remained controversial, resulting in the stalling of any therapeutic perspective. Due to the supposed cell adhesive properties of CD31, specific monoclonal antibodies and recombinant proteins were regarded as blocking agents because their use prevented the arrival of leukocytes at sites of acute inflammation. However, the observed effect of those compounds likely resulted from the engagement of the immunomodulatory function of CD31 signaling. This was acknowledged only later though, upon the discovery of CD31’s 2 intracytoplasmic tyrosine residues called immunoreceptor tyrosine inhibitory motifs. A growing body of evidence currently points at a therapeutic potential for CD31 agonists in atherothrombosis. Clinical observations show that CD31 expression is altered at the surface of leukocytes infiltrating unhealed atherothrombotic lesions and that the physiological immunomodulatory functions of CD31 are lost at the surface of blood leukocytes in patients with acute coronary syndromes. On the contrary, translational studies using candidate therapeutic molecules in laboratory animals have provided encouraging results: synthetic peptides administered to atherosclerotic mice as systemic drugs in the acute phases of atherosclerotic complications favor the healing of wounded arteries, whereas the immobilization of CD31 agonist peptides onto coronary stents implanted in farm pigs favors their peaceful integration within the coronary arterial wall.
Article
Full-text available
Background: Previous studies have shown that hyperbaric oxygen therapy (HBOT) can improve the motor functions and memory of post-stroke patients in the chronic stage. Objective: The aim of this study is to evaluate the effects of HBOT on overall cognitive functions of post-stroke patients in the chronic stage. The nature, type and location of the stroke were investigated as possible modifiers. Methods: A retrospective analysis was conducted on patients who were treated with HBOT for chronic stroke (>3 months) between 2008-2018. Participants were treated in a multi-place hyperbaric chamber with the following protocols: 40 to 60 daily sessions, 5 days per week, each session includes 90 min of 100% oxygen at 2 ATA with 5 min air brakes every 20 minutes. Clinically significant improvements (CSI) were defined as > 0.5 standard deviation (SD). Results: The study included 162 patients (75.3% males) with a mean age of 60.75±12.91. Of them, 77(47.53%) had cortical strokes, 87(53.7%) strokes were located in the left hemisphere and 121 suffered ischemic strokes (74.6%).HBOT induced a significant increase in all the cognitive function domains (p < 0.05), with 86% of the stroke victims achieving CSI. There were no significant differences post-HBOT of cortical strokes compared to sub-cortical strokes (p > 0.05). Hemorrhagic strokes had a significantly higher improvement in information processing speed post-HBOT (p < 0.05). Left hemisphere strokes had a higher increase in motor domain (p < 0.05). In all cognitive domains, the baseline cognitive function was a significant predictor of CSI (p < 0.05), while stroke type, location and side were not significant predictors. Conclusions: HBOT induces significant improvements in all cognitive domains even in the late chronic stage. The selection of post-stroke patients for HBOT should be based on functional analysis and baseline cognitive scores rather than the stroke type, location or side of lesion.
Article
Full-text available
The best known form of oxygen therapy is hyperbaric oxygen (HBO) therapy, which increases both concentration and atmospheric pressure. HBO supports tissue regeneration and is indicated in an increasing number of pathologies. Less known but still showing some promising effects is normobaric oxygen (NBO) therapy, which provides some advantages over HBO including eliminating barotrauma risk, increased ease of administration and a significant cost reduction. However, still little is known about differences and similarities in treatment effects between HBO and NBO. Therefore we tested whether NBO induces a biological response comparable to HBO with a focus on stem progenitor cell mobilization and changes in serum cytokine concentration. We randomly assigned Sprague-Dawley rats into an NBO treatment group (n = 6), and a room air control group (n = 6). The NBO treatment group was exposed to 42% oxygen for 2 hours a day for 10 days. The room air group was concurrently kept at 20.9% oxygen. The frequency and number of stem progenitor cells in peripheral blood were analyzed by flow cytometry. Plasma cytokine expression was analyzed by cytokine array enzyme linked immunosorbent assay. All analyses were performed 24 hours after the final exposure to control for transient post treatment effects. The NBO treatment group showed an increase in circulating CD133+/CD45+ stem progenitor cell frequency and number compared to the room air control group. This rise was largely caused by CD34- stem progenitor cells (CD133+/CD34-/CD45+) without changes in the CD34+ population. The plasma cytokine levels tested were mostly unchanged with the exception of tumor necrosis factor-α which showed a decrease 24 hours after the last NBO exposure. These findings support our hypothesis that NBO induces a biological response similar to HBO, affecting serum stem progenitor cell populations and tumor necrosis factor-α concentration. The study was approved by Institutional Animal Care and Use Committee (IACUC) of the University of Wisconsin, Madison, WI, USA (approval No. M005439) on June 28, 2016.
Article
Endothelial progenitor cells (EPCs) promote angiogenesis and play an important role in myocardial and vascular repair after ischemia and infarction. EPCs consist of different subpopulations including CD34‑CD133+ EPCs, which are precursors of more mature CD34+CD133+ EPCs and functionally more active in terms of homing and endothelial regeneration. In the present study we analyzed the functional and differentiation abilities of CD34‑CD133+ EPCs. Isolation of EPC populations (CD34+CD133+, CD34‑CD133+) were performed by specific multi‑step magnetic depletion. After specific stimulation a significant higher adhesive and migrative capacity of CD34‑CD133+ cells could be detected compared to CD34+CD133+ cells (P<0.001, respectively). Next to this finding, not only significantly higher rates of proliferation (P<0.005) were detected among CD34‑CD133+ cells, but also a higher potential of cell‑differentiation capacity into other cell types. Next to a significant increase of CD34‑CD133+ EPCs differentiating into a fibroblast cell‑type (P<0.001), an enhancement into a hepatocytic cell‑type (P=0.033) and a neural cell‑type (P=0.016) could be measured in contrast to CD34+CD133+ cells. On the other hand, there was no significant difference in differentiation into a cardiomyocyte cell‑type between these EPC subpopulations (P=0.053). These results demonstrate that EPC subpopulations vary in their functional abilities and, to different degrees, have the capacity to transdifferentiate into unrelated cell‑types such as fibroblasts, hepatocytes, and neurocytes. This indicates that CD34‑CD133+ cells are more pluripotent compared to the CD34+CD133+ EPC subset, which may have important consequences for the therapy of vascular diseases.