Content uploaded by Ján Mašán
Author content
All content in this area was uploaded by Ján Mašán on May 16, 2021
Content may be subject to copyright.
1
REVIEW ARTICLE
THE POSSIBILITIES OF USING THE EFFECTS OF OZONE THERAPY IN
NEUROLOGY
Ján MASAN1,2,3, Miron SRAMKA2 , Daria RABAROVA3
1 University of St. Cyril and Methodius in Trnava, Slovak Republic.
2 St. Elizabeth University of Health Care and Social Work in Bratislava, Slovak Republic.
3 Trnava University in Trnava, University Hospital Trnava, Slovak Republic.
Correspondence to: h.Doc. MUDr. Ján Mašán, PhD.
St. Elizabeth University of Health Care and Social Work in Bratislava, Slovak Republic
E-MAIL : masanjan@gmail.com
Keywords: Ozone therapy. Major ozone therapy. Neurodegenerative disorders. Antioxidant
system. Oxidative stress biomarkers. Multiple sclerosis. Stroke. Neuropathy. Phantom limb
pain. Polyneuropathy.
--------------------------------------------------------------------------------------------------------
Abstract
OBJECTIVES: The beneficial effects of ozone therapy consist mainly of the promotion of
blood circulation: peripheral and central ischemia, immunomodulatory effect, energy boost,
regenerative and reparative properties, and correction of chronic oxidative stress. Ozone
therapy increases interest in new neuroprotective strategies that may represent therapeutic
targets for minimizing the effects of oxidative stress.
METHODS: The overview examines the latest literature in neurological pathologies treated
with ozone therapy as well as our own experience with ozone therapy. The effectiveness of
treatments is connected to the ability of ozone therapy to reactivate the antioxidant system to
address oxidative stress for chronic neurodegenerative diseases, strokes, and other
pathologies. Application options include large and small autohemotherapy, intramuscular
application, intra-articular, intradiscal, paravertebral and epidural, non-invasive rectal,
transdermal, mucosal, or ozonated oils and ointments. The combination of different types of
ozone therapy stimulates the benefits of the effects of ozone.
2
RESULTS: Clinical studies on O2-O3 therapy have been shown to be efficient in the
treatment of neurological degenerative disorders, multiple sclerosis, cardiovascular, peripheral
vascular, orthopedic, gastrointestinal and genitourinary pathologies, fibromyalgia, skin
diseases/wound healing, diabetes/ulcers, infectious diseases, and lung diseases, including the
pandemic disease caused by the COVID-19 coronavirus.
CONCLUSION: Ozone therapy is a relatively fast administration of ozone gas. When the
correct dose is administered, no side effects occur. Further clinical and experimental studies
will be needed to determine the optimal administration schedule and to evaluate the
combination of ozone therapy with other therapies to increase the effectiveness of treatment.
Abbreviations: O3 – ozone, GSH – glutathione, NADPH – nicotinamide adenine
dinucleotide, (SOD) – superoxide dismutase, PUFA – unsaturated fatty acids, LOP – lipid
ozonation products, ARE – antioxidant response elements, ATP – adenosine triphosphate,
ROS – reactive oxygen species, DNA – deoxyribonucleic acid, NADH – nicotinamide
adenine dinucleotide
INTRODUCTION
Van Mauren was first to discover the distinctive odor of O3 in 1785 (Altman 2007).
However, its first identification as a distinct chemical compound was made by Schönbein in
Basel in 1840. In 1896, Nikola Tesla patented a generator for producing ozone. The use of
ozone became normal practice after the studies of Dr. H.H. Wolff. In 1915, during World War
I, ozone was used to treat war wounds. Following Bocci’s studies, ozone therapy has been
incorporated into the treatment of chronic inflammatory diseases in the orthopedic field.
When used in appropriate doses, it has been shown to be effective in inducing well-tolerated
oxidative stress. Ozone is a metastable substance and must be generated on-site.
Contraindication of O3 therapy is lung inhalation, activating factors triggering an
inflammatory response (Bocci 2006; Pryor et al. 2019). Jacobs (1982) carefully examined all
the possible negative effects of ozone therapy. Despite the famous “toxicity” of ozone, it
appears that the incidence is only 0.0007%, one of the lowest in medicine. Four deaths due to
direct IV injection of the gas were included in his data but, since 1982, other deaths due to
malpractice have occurred, of which at least three were in Italy (Bocci 2010).
3
It is currently applied in a wide range of medical fields: surgery, orthopedics,
neurology, oncology, dermatology, cardiology, psychiatry, radiology, rheumatology,
gynecology, gastroenterology, angiology, dentistry, etc.
The effects of medical ozone as a substance can be attributed to those defined by
hormesis (from the Greek word hormáein = to set in motion), i.e. referring to the hypothesis
regarding the beneficial effects of low doses. At the same time, it triggers a response with
high doses, thus increasing the body’s resistance.
Induction of the response to stress by short low doses usually protects the body for a
longer period of time and against other possible types of doses (Rattan et al. 2009). The
administration of O3 therapy varies based on treatment goals and treatment focus. Ozone
therapy combines a mixture of oxygen (O2)-O3 with a diverse therapeutic range (10–80
μg/ml of gas per ml of blood) (Bocci 2006). Human blood contains a large number of
antioxidants such as uric acid, ascorbic acid, cysteine, glutathione, albumin, certain chelating
proteins such as albumin, and enzymes such as catalase, the redox system of GSH, NADPH
(nicotinamide adenine dinucleotide), and superoxide dismutase (SOD) (Bocci et al., 2005).
Like any other gas, ozone dissolves upon contact with body fluids according to
Henry’s Law in relation to temperature, pressure, and ozone concentration. It reacts
immediately as soon as it is dissolved: O3 + biomolecules → O2 + O2 + energy. Atomic
oxygen behaves as a reactive atom. The fundamental ROS (reactive oxygen species) molecule
is hydrogen peroxide H2O2, which is a non-radical oxidant able to act as an ozone Messenger
responsible for eliciting several biological and therapeutic effects. In physiological amounts,
they act as regulators of signal transduction and represent important mediators of host defense
and immune responses (Dattilo et al. 2015).
The reaction of O3 with water causes the formation of one mole of hydrogen peroxide
(H2O2) and two moles of lipid oxidation products with polyunsaturated fatty acids (PUFA),
forming a mixture of lipid ozonation products (LOP) (Barone et al. 2015) including
lipoperoxyl radicals (Inal et al. 2011). Moderate oxidative stress caused by O3 increases the
activation of the transcription factor of the pathway-mediated nuclear factor-related erythroid
factor 2 (Nrf2) (Bocci et al. 2015, Re et al. 2014), which is responsible for activating the
transcription of antioxidant response elements (ARE). After their induction, the concentration
of antioxidant enzymes increases in response to the transient oxidative stress O3 (Bocci et al.
2015).
4
POTENTIAL APPLICATION OF OZONE THERAPY
Vascular and Haematological Modulation The production of antioxidant enzymes
affects the whole body (Gonzalez et al. 2004; Valacchi 2000). O3 is a stimulator of O2
transmembrane flow. Increased intracellular O2 levels secondarily form the mitochondrial
respiratory chain (Madej et al. 2007). In red blood cells, O3 increases phosphofructokinase
activity, thereby increasing the rate of glycolysis. Increased glycolytic rate increases ATP and
2,3-diphosphoglycerate (2,3-DPG) in the cell. Following a long treatment cycle of the elderly,
Bocci et al. 2011 noted a significant increase in the 2,3-diphosphoglycerate level in
oxyhaemoglobin. As a result of the Bohr effect, its dissociation curve shifts to the right,
making it easier to transfer oxygen. Under physiological conditions, the endothelium regulates
vascular tone (Molinari et al. 2017). Ozonised blood increases the release of prostacyclin
(PGI2) and angiopoietins, both important factors in improving ischemic vasculopathy
(Fernández et al. 2008; Elvis et al. 2011).
The correction of chronic oxidative stress via the increase of antioxidant enzymes can
increase erythroblast differentiation. This leads to a progressive increase in erythrocytes and
preconditions them to having resilience towards oxidative stress, to an increase of
erythrocytes with improved metabolic properties, as well as ensuring that young erythrocytes
contain more G6PDH than older cells generated prior to the treatment, which is a type of
“super-gifted erythrocytes” capable of correcting hypoxia in vascular diseases (Chang et al.
2005). An improvement in blood circulation and oxygen supply to ischemic tissues has been
recorded, leading to an increased supply of O2 to hypoxic tissues (Brigelius-Flohé et al. 2011).
Activation of the Immune System. O3 has been shown to react with antioxidants and
alter peroxidation compounds. H2O2 has been shown to act as a regulatory step in signal
transduction by diffusing into immune cells and by facilitating a myriad of immune responses
(Gulmen et al. 2013; Caliskan et al. 2011). An increase in interferon, tumor necrosis factor,
and interleukin (IL)-2 was observed. IL-2 increases were initiated by immune response
mechanisms (Elvis 2011). In addition, H2O2 activates nuclear factor-kappa B (NF-κB) and
transforms growth factor-beta (TGF-β), thus increasing the immunoactive release of cytokines
and tissue refurbishment.
After each restarted therapy, a small percentage of immune cells are activated and
these cells release cytokines into the micro-environment, activating neighboring cells and, as
a result, slowly enhancing immune responses. Only submicromolar concentrations can reach
5
all organs, particularly bone marrow, liver, central nervous system, endocrine glands, etc.,
where they act as signaling molecules of an ongoing acute oxidative stress (Mancuso et al.
1997). These molecules can elicit the upregulation of antioxidant enzymes. The induction of
HO-1 has been described as one of the most important antioxidant defense and protection
enzymes. Throughout the treatments, LOP acts as an acute oxidative stressor in the bone
marrow micro-environments and thus activates the release of metalloproteinases, of which
particularly MP-9 may favor the detachment of staminal cells. These cells, once in the blood
circulation, may be attracted to sites where a previous injury has taken place (Mancuso et al.
2008).
Lahodny (2021) collected ten blood samples with 70 μg/ml of ozone and reinfusion from a
patient as part of a session to implement the therapeutic concept. This therapy generated a
significant activation of stem cells, and subsequent rapid healing of wounds and inflammation
were documented in patients (Rowen 2018). Thanks to this therapy, a several-year defect was
cured in a mere 14 days (Mašán 2018).
Ozone therapy has a neuroimmunomodulatory effect, it activates the psychosomatic
system, thus allowing the release of the growth hormone ACTH-cortisol, neurotonic
hormones, and neurotransmitters. We clarified why patients report a feeling of euphoria and
wellness during therapy: the disappearance of asthenia and depression, a reduction of
pessimism syndrome, associated with a lack of side effects, represent positive results
(Fernández et al. 2008; Mancuso 2017).
CLINICAL APPLICATION OF OZONE THERAPY IN NEUROLOGY
The first beneficial effects of ozone in the treatment of neurological disorders refer to the
treatment of headaches and facial pain associated with pathological changes in the optic
thalamus. Ozone is used to treat allodynia, neuropathic pain, and hyperalgesia (Kal et al.
2017; Hu et al. 2018).
Neurodegenerative diseases – Parkinson’s disease, Alzheimer’s and Wilson’s disease, senile
and vascular dementia, amyotrophic lateral sclerosis, optic nerve dysfunction, bilateral
sensorineural hearing loss, and maculopathy, Huntington’s disease, cognitive and movement
disorders of the elderly who experience common effects of oxidative stress (Aso et al. 2012).
The process of aging is characterized by the loss of homeostasis, leading to pathologic
6
formation of reactive oxygen species (ROS), mitochondrial dysfunction, and metabolic
unbalance (Dugger et al. 2017). These pathophenotypes determine abnormal aggregation of
specific proteins (Yanar et al. 2020), given the connection between excessive ROS
accumulation and impairment in the proteostasis network. A natural bioactive molecule with
an antioxidant property such as ozone (O3) can be indicated as a potential new strategy to
delay neurodegeneration. This hypothesis is based on the evidence regarding the interaction
between O3 and Nrf2 (Galie et al. 2018; Siniscalco et al. 2018; Re et al. 2014; Vaillant et al.
2013). Molecular mechanisms related to antioxidant/anti-apoptotic/pro-autophagy processes
targeted by O3 administration via an Nrf2 biological pathway.
It is best to implement ozone therapy (O2-O3) in an early phase before the potential
development of a neurodegenerative pathology (Scassellati et al. 2020). The O2 availability
affects the expression of different hypoxia-inducible factors (HIFs) and plays the role of a
cellular adapter to hypoxia (Curro et al. 2018; Zhang et al. 2014; Re et al. 2014), leading to
the activation of trophic proteins and, consequently, to specific biological processes, including
erythropoiesis and angiogenesis (Zhou et al. 2019).
Antioxidant Property of Ozone (O3): Oxidative stress is a condition where ROS production
exceeds the cellular antioxidant defense system, leading to an imbalance between the two
systems, and this may contribute to neuronal damage. It has implications on the pathogenesis
and progression of neurodegenerative diseases (Singh et al. 2019).
Oxidative damage may impair the cells in their structure and function as a result of the
reduced activity of mitochondria. The damage is not confined to the brain but is also evident
in peripheral cells and tissues, which require a high-energy source such as the heart, muscles,
brain, or liver. To function properly, neurons rely on the mitochondria, which produce the
energy required for most of the cellular processes, such as neurotransmitter synthesis,
including synaptic plasticity.
Mitochondrial dysfunctions cause an increase in ROS for lowered oxidative capacity and
antioxidant defense, resulting in increased oxidative damage to protein and lipids, decreased
ATP production, and accumulation of DNA damage. Moreover, mitochondrial bioenergetic
dysfunction and the release of pro-apoptotic mitochondrial proteins into the cytoplasm initiate
a variety of cell death pathways (Garcia-Escudero et al. 2013; Reutzel et al. 2020).
Ozone therapy stimulates the Krebs cycle by enhancing the oxidative carboxylation of
pyruvate and stimulating the production of adenosine triphosphate (ATP) (Guven et al. 2008).
7
It causes a significant reduction of nicotinamide adenine dinucleotide (NADH), an increase of
the coenzyme A levels to fuel the Krebs cycle, and oxidizes cytochrome C (Brigelius-Flohe et
al. 2011; Elvis et al. 2011).
Ozone(O3)-influenced pro-oxidative and antioxidant defense biomarkers and their role in
aging processes and neurodegenerative disorders (ND). Twenty-nine biomarkers implicated in
oxidative stress, in endogenous antioxidant, and in vitagene systems have been identified.
These biomarkers have been studied and found modulated after O3 therapy performed in in
vivo (human and animal models) samples. Neurodegenerative disorders are characterized by
progressive loss of cognitive and behavioral deterioration (Scassellati et al. 2020).
Molecular Mechanisms Involving Ozone Therapy and Their Biological Relevance in
Neuroprotection of Nrf2, and the Vitagene Network. Oxidative stress is a condition where
ROS and nitrogen (RNS) production exceeds the cellular antioxidant defense system, leading
to an imbalance between the two systems and this may contribute to neuronal damage and
abnormal neurotransmission. ROS and RNS are also major factors in cellular senescence that
leads to an increase in the number of senescent cells in tissues on a large scale (Liguori et al.
2018). Cellular senescence is a physiological mechanism that stops cellular proliferation in
response to damage that occurs during replication. Senescent cells acquire an irreversible
senescence-associated secretory phenotype (SASP), involving secretion of soluble factors
(interleukins, chemokines, and growth factors), degradative enzymes such as matrix
metalloproteases (MMPs), and insoluble proteins/extracellular matrix (ECM) components.
When O3 is administrated, it dissolves immediately in the plasma/serum and it reacts with
PUFA (polyunsaturated fatty acids), leading to the formation of the two fundamental
messengers: hydrogen peroxide (H2O2) as a ROS and 4-hydroxynonenal (4HNE) as a lipid
oxidation product (LOP) (Bocci et al. 1998).
LOPs diffuse into all cells and inform them of minimal oxidative stress. After the
oxidative/electrophilic stress challenge (Ishii et al. 2004), other aldehydes (Levonen et al.
2004) induced by O3 (Galie et al. 2018, Siniscalco et al. 2018, Re et al. 2014, Vaillant et al.
2013) inhibit the Nrf2 conjugation, provoking the nuclear accumulation of Nrf2. This leads to
the decreased expression of pro-inflammatory cytokines.
Another mechanism involves casein kinase 2 (CK2), another regulator of Nrf2 activity
through its phosphorylation. It has been demonstrated that O3 influences CK2 levels together
with Nrf2 phosphorylation, reducing oxidative stress and pro-inflammatory cytokines in
multiple sclerosis patients (Delgado-Roche et al. 2017). Similarly, O3 inhibits oxidative stress
8
through inhibition of the mitogen-activated protein kinase phosphatase (MAPK) 1 signaling
pathway (Wang et al. 2018a).
Oxidative stress is one of the major drivers of protein misfolding that, accumulating and
aggregating as insoluble inclusions, can determine neurodegeneration (Hohn et al.
2020; Knowles et al. 2014). It is known that Nfr2 promotes the clearance of oxidized or
otherwise damaged proteins through the autophagy mechanism (Tang et al. 2019),
Interestingly, O3 can also modulate the degradation protein systems, not only via the Nrf2
pathway but also via activation of the AMP-activated protein kinase (AMPK)/mammalian
target of rapamycin (mTOR) signaling pathway, as demonstrated (Zhao et al. 2018).
O3 can protect against the overproduction of nitric oxide (NO) when NO is a toxic oxidant.
NO can rapidly react with other free radicals such as O2 − the highly reactive oxidant
peroxynitrite (ONOO) - and other RNS, which in turn damage the biomolecules (e.g. lipids,
protein, DNA/RNA), playing a key role in chronic inflammation and neurodegeneration
(Massaad, 2011; Toda et al. 2009), It has been demonstrated that O3 downregulates inducible
nitric oxide synthase (iNOS), which generates NO (Manoto et al. 2018; Smith et al.
2017) via NF-κB signaling.
CO acts as an inhibitor of another important pathway, NF-κB (nuclear factor kappa B subunit
1) signaling, which leads to the decreased expression of pro-inflammatory cytokines, while
bilirubin also acts as an important lipophilic antioxidant. Furthermore, HO-1 directly inhibits
pro-inflammatory cytokines and activates anti-inflammatory cytokines, leading to a balancing
of the inflammatory process (Ahmed et al. 2017). Our research group confirmed that mild
ozonization, tested on in vitro systems, induced modulation of genes including HO-1
(Scassellati et al. 2017),
In addition, Nrf2 also regulates the constitutive and inducible expression of antioxidants
including, but not limited to, Superoxide Dismutases (SOD), Glutathione Peroxidase (GSH-
Px), Glutathione-S-Transferase (GST), Catalase (CAT), and NADPH quinone oxidoreductase
1 (NQO1), phase II enzymes of drug metabolism and HSP (Galie et al. 2018, Bocci et al.
2015, Pedruzzi et al. 2012).
In this context, Nrf2 is considered a hormetic-like pathway (Calabrese et al. 2010). It
has widely been reported that the activation of Nrf2 by several different mechanisms (calorie
9
restriction, physical exercise, polyphenols) can be a way to improve health due to its
transcriptional modulation on the vitagene network. Nfr2 is strongly implicated in aging
processes (Zhang et al. 2015; Schmidlin et al. 2019; Silva-Palacios et al. 2018). These
conditions share common mechanisms, and the results represent a first attempt to structure
Nrf2 as a common therapeutic medicine approach. Research on the antioxidant activities of
O3 correlated with the interaction with Nrf2 (Galie et al. 2018; Siniscalco et al. 2018; Re et al.
2014; Vaillant et al. 2013). Different antioxidants can combat many associated pathologies,
including neurodegenerative disorders (Leri et al. 2020; Calabrese 2020). The mechanisms of
the positive effects of O3 are attributed not only to up-regulation of cellular antioxidant
enzyme activity, but also to the activation of the immune and anti-inflammatory systems,
modulation of NPRL3 inflammasome, action on the proteasome, enhancement in the release
of growth factors from platelets, improvement in blood circulation and O2 delivery to
damaged tissues, and enhancement of general metabolism (Scassellati et al. 2020).
The gradual escalation of the ozone dose enhances cerebral blood flow, improves metabolism,
and corrects chronic oxidative stress. Neuronal cells may reactivate the synthesis of
antioxidant enzymes, which is crucial to normalize the redox state and avoid cell death. The
local induction of haeme oxygenase-1 played a critical role in reducing oxidative damage.
The enzyme caused the local release of CO and bilirubin that acts as a potent antioxidant of
peroxynitrite (Clavo et al. 2004). The trace amounts can pass through the blood-brain barrier
to reach the sites of neurodegeneration and upregulate the cellular synthesis of antioxidant
enzymes, which is a crucial step towards readjusting the impaired cell redox system.
Neurodegenerative disorders affect approximately 50 million people globally and have a
terrific and increasingly negative social-economic impact on families and society. A better
understanding of degenerative events and the effects of ozone therapy during the early stage
of the disease may be able to slow down the demise of critical populations of neurons and
thus provide patients with a better quality of life through ozone therapy. The use of ozone in
the treatment of various diseases can have a therapeutic effect, provided that the correct dose
is administered at the right time interval and depending on the antioxidant capacity of the
tissue exposed, as well as ensuring the concentration is used within a non-toxic range. The
versatility of ozone therapy is due to the cascade of ozone-derived compounds able to act on
several targets leading to a multifactorial correction of various pathological conditions. Ozone
therapy can improve well-being and delay the negative effects of aging. The aging process is
essentially linked to the balance of oxidants and antioxidants, advanced glycation end
10
products (AGE), the role of genes and the immune system, the importance of telomeres and
telomerase, hormones, nutrition, environmental factors, and certain other factors. Stem cell
therapy, virtual reality rehabilitation, electromagnetic fields, and ozone therapy are among the
therapeutic approaches in the treatment of neurodegenerative disorders (Mitrečić et al. 2020).
Ozone Autohaemotherapy Induces Cerebral Metabolic Changes in Multiple Sclerosis
Patients.
The oxygenated hemoglobin concentration is increased and the chronic oxidative stress level
typical of MS sufferers is reduced (Molinari et al. 2014). Ozone has an effect on biomarkers
of oxidative stress and inflammation, it significantly improves the activity of antioxidant
enzymes and increases the reduced cellular glutathione levels, and demonstrates antioxidant
and anti-inflammatory effects.
Ozone promotes Nrf2 phosphorylation, reducing oxidative stress and pro-inflammatory
cytokines in multiple sclerosis patients (Delgado-Rosche et al. 2017).
Mechanisms of vascular pathophysiology in multiple sclerosis patients treated with ozone
therapy demonstrate revascularisation and regeneration of the blood-brain barrier as a result
of immunoreactive glial cells coming into contact with blood vessel walls during ozone
therapy (Ameli et al. 2019; Biomed 2019). The cerebrovascular system enhanced by ozone
autohemotherapy in multiple sclerosis patients increases brain metabolism and helps them
recover from the lower activity levels that predominate in MS patients (Molinari et al. 2017).
At both primary and chronic stages of MS, antioxidant therapy is an active approach to
disease progression. O3 therapy increases total tissue-oxygen levels in patients with multiple
sclerosis. Ozone therapy is a therapeutic alternative for patients with multiple sclerosis
(Molinari et al. 2014; Simonetti et al. 2014).
Ozone therapy applied in acute ischemic stroke increases blood oxygen saturation,
improves blood circulation, activates erythrocyte metabolism, improves tissue oxygenation
and oxygen supply, restores cell function, promotes oxygen metabolism, and induces
thrombolysis by hydrogen peroxide (H2O2) formation (Qiu et al. 2021). Ozone was
administered daily via rectal inflation (at an ozone concentration of 40 mg/l and 200 ml) for
15 days. In the clinical phase, an improvement in the quality of life was recorded in 80% of
patients treated with ozone therapy Qiu et al. 2021). Ozone therapy has certain therapeutic
benefits for ischemic stroke patients. Jing Qiu, Hui-sheng Chen (2016) reported that ozonated
autohemotherapy substantially improved neurological function in stroke patients.
11
Neurological Deficits Treated with Ozone Therapy. Trigeminal neuralgia, postherpetic
neuralgia, meningitis, cervical myelopathy, demyelinating of nerves. Acoustic neuroma,
muscular neuropathy, and parkinsonism have shown satisfactory improvements
(Kakkad 2018).
The treatment of trigeminal neuralgia with percutaneous ozone and injections into the
Gasserian ganglion has been shown to have positive long-term effects on pain (An et al. 2018;
Gao et al. 2020).
The treatment of phantom limb pain with ozone injection into the nerve root in the stump
was met with positive results (Li et al. 2020).
Ozone therapy in patients with pain secondary to chemotherapy-induced peripheral
neuropathy with cancer of the colon and rectum treated with oxaliplatin and rectal
insufflation sessions of O3/O2 as part of the ongoing randomized controlled trial
(O3NPIQ) (O3NPIQ) 2020 (Clavo et al. 2019). Chemotherapy-induced peripheral
neuropathy decreases the quality of life of patients and can lead to a decrease in and
interruption of chemotherapy treatment. Potential pathophysiological mechanisms involved in
chemotherapy include chronic oxidative stress and consequent increases in free radicals and
pro-inflammatory cytokines. Several antioxidant‐based therapies have been tested. On the
other hand, ozone therapy can elicit an adaptive antioxidant and anti-inflammatory response
that could prove to be useful (Clavo et al. 2021).
Ozone therapy in cerebral ischemia and hypometabolism in meningioma treated by
stereotactic radiosurgery. Following ozone autohemotransfusion treatment, there was an
improvement in brain perfusion and metabolism, as demonstrated by SPECT and PET scans
(Clavo et al. 2011). Rectal ozone therapy showed positive effects of improving
neurorehabilitation in children with infratentorial ependymoma (Mašán & Golská 2017).
Ozone therapy of resistant meningitis in infants – a mixture of ozone gas with pure oxygen
was used to treat resistant meningitis in infants with hydrocephalus and the infection was
cured (Dahhan 2015).
Endolumbal ozone therapy in the treatment of patients with a complicated spinal injury
in the acute period improved neurological symptoms (Yuldashev et al. 2020).
In the treatment of lumbar discopathy, ozone therapy is used with verified clinical benefits
in the treatment of lumbar disc damage. Ozone application methods can be administered
12
paravertebrally, juxtaforaminally, intradiscally (De Oliveira – Magalhaes et al. 2012; Mašán
2017; Li et al. 2020; Yuldashev et al. 2020).
CONCLUSION
Ozone therapy is a biological treatment method with a wide range of applications in
medicine. The versatility of ozone therapy is due to the cascade of ozone-derived compounds
able to act on several targets leading to a multifactorial correction of pathological conditions.
O3 therapy induces moderate oxidative stress when interacting with lipids, increases
endogenous production of antioxidants, local perfusion, and oxygen delivery, as well as
enhances immune responses. Oxidative stress occurs when there is an imbalance between free
radical formation and antioxidant defense and is associated with damage to lipids, proteins,
and nucleic acids. Ozone is being examined as a master regulator of multiple cytoprotective
responses, as a key actor across a wide range of diseases, and as a therapeutic objective for
aging and aging-associated disorders. It provides scientific evidence for the application of
oxygen-ozone (O2-O3) in the treatment of neurological diseases. Oxidative stress is currently
thought to play a significant role in the development of inflammatory diseases, ischemic
diseases, hypertension, Alzheimer’s disease, Parkinson’s disease, muscular dystrophy, and
many others.
The versatility of ozone therapy is due to the cascade of ozone-derived compounds able to act
on several targets leading to a multifactorial correction of various pathological conditions, as
well as cardiovascular, peripheral vascular, neurological, orthopedic conditions, skin diseases,
wound healing, diabetes, and lung diseases, including the pandemic disease caused by the
COVID-19 coronavirus. Ozone therapy also promotes tissue perfusion, immunomodulatory
effect, energy effect of the body, and has regenerative and reparative properties. It appears to
be a potentially effective treatment method. When the correct dose is administered, no side
effects occur. Further clinical and experimental studies will be needed to determine the
optimal administration schedule and to evaluate the potential combination of ozone therapy
with other therapies to increase the effectiveness of treatment.
REFERENCES
1. Ademowo OS, Dias HKI, Milic I, Devitt A, Moran R, Mulcahy R, Howard AN, Nolan
M, Griffiths HR. (2017). Phospholipid oxidation and carotenoid supplementation in
13
Alzheimer’s disease patients. Free Radic. Biol. Med. 108: 77–85. doi: 10.1016 /
j.freeradbiomed.2017.03.008.
2. Ahmed SM, Luo L, Namani A, Wang XJ, Tang X. (2017). Nrf2 signaling pathway:
Pivotal roles in inflammation. Biochim Biophys Acta Mol Basis Dis. 1863(2): 585-
597. doi: 10.1016/j.bbadis.2016.11.005.
3. Altman N. Healing Arts (2007). The oxygen prescription : the miracle of oxidative
therapies. Healing Arts Press, Rochester, Vermont 2007. ISBN1-59477- 4.
4. Ameli J, Banki A, Khorvash F, Simonetti V, Jafari NJ, Izadi M (2019). Mechanisms of
pathophysiology of blood vessels in patients with multiple sclerosis treated with ozone
therapy: a systematic review. Acta Biomed. 90: 213–217. doi:
10.23750/abm.v90i3.7265.
5. An JX, Liu H, Chen RW, Wang Y, Zhao WX, Eastwood D, Williams JP (2018).
Computed tomography-guided percutaneous ozone injection of the Gasserian ganglion
for the treatment of trigeminal neuralgia. 11: 255-263. doi.org/10.2147/JPR.S140369.
6. Aso E, Lomoio S, Lopez-Gonzalez I, Joda L, Carmona M, Fernandez-Yague N,
Moreno J, Juves S, Pujol A, Pamplona R, Portero-Otin M, Martin V, Diaz M, Ferrer I
(2012). Amyloid generation and dysfunctional immunoproteasome activation with
disease progression in animal model of familial Alzheimer’s disease. Brain Pathol.
22: 636–653. doi: 10.1111 / j.1750-3639.2011.00560.x.
7. Barone P, Santangelo G, Morgante L ,Onofrj M, Meco G, Abbruzzese G,
Bonuccelli U, Cossu G , Pezzoli G , Stanzione P, Lopiano L , Antonini A , Tinazzi
M. (2015). A randomized clinical trial to evaluate the effects of rasagiline on
depressive symptoms in non-demented Parkinson's disease patients. Eur J Neurol.
22(8): 1184-91. doi: 10.1111/ene.12724.
8. Bilge A, Ozturk O, Adali Y, Ustebay S. (2018). Could Ozone Treatment be a
Promising Alternative for Osteomyelitis? an Experimental Study. Acta Ortop. Bras.
26: 67–71. doi: 10,1590 / 1413-785220182601179926.
9. Biomed A (2019). Mechanisms of pathophysiology of blood vessels in patients with
multiple sclerosis treated with ozone therapy: a systematic review . 90 (3): 213–217.
doi: 10.23750/abm.v90i3.7265.
10. Bocci V (2010). The Potential Toxicity of Ozone: Side Effects and Contraindications
of Ozonetherapy. OZONE. 75-84. doi:10.1007/978-90-481-9234-2_7.
11. Bocci V, Larini A, Micheli V. (2005). Restoration of normoxia by ozone therapy may
control neoplastic growth: a review and a working hypothesis. J Altern Complement
Med. 11(2): 257-65. doi: 10.1089/acm.2005.11.257.
12. Bocci V, Valacchi G (2015). Nrf2 activation as target to implement therapeutic
treatments. Frontiers in Chemistry. 3: 4. doi: 10.3389/fchem.2015.00004.
13. Bocci V, Valacchi G, Corradeschi F, Fanetti G. (1998). Studies on the biological
effects of ozone: 8. Effects on the total antioxidant status and on interleukin-8
production. Mediators Inflamm. 7(5): 313–317.doi: 10.1080/09629359890820.
14. Bocci V, Zanardi I, Huijberts MS, Travagli V. (2011). Diabetes and chronic oxidative
stress. A perspective based on the possible usefulness of ozone therapy. Diabetes
Metab Syndr. 5: 45–49. doi: 10.1016 / j.dsx.2010.05.014.
15. Bocci VA (2006). Tropospheric ozone toxicity vs. usefulness of ozone therapy. Arch
Med Res 2007, Feb; 38(2): 265-267. doi: 10.1016/j.arcmed.2006.09.011.
16. Bocci VA, Zanardi I, Travagli V. (2011). Ozone acting on human blood yields a
hormetic dose-response relationship. J Transl Med. 9: 66. doi: 10,1186 / 1479-5876-
9-66.
17. Bocci VA. (2006) Scientific and Medical Aspects of Ozone Therapy. State of the Art.
Archives of Medical Research, 37, 425-435. doi.org/10.1016/j.arcmed.2005.08.006
14
18. Braidy N, Izadi M, Sureda A, Jonaidi-Jafari N, Banki A, Nabavi SF, Nabavi S.M
(2018). Therapeutic relevance of ozone therapy in degenerative diseases: Focus on
diabetes and spinal pain. J. Cell. Physiol. 233: 2705–2714. doi: 10.1002/jcp.26044.
19. Brigelius-Flohe R, Flohe L. (2011). Basic principles and emerging concepts in the
redox control of transcription factors. Antioxid.Redox Signal. 15: 2335–2381. doi:
10.1089/ars.2010.3534.
20. Calabrese EJ. (2020). Hormesis and Ginseng: Ginseng Mixtures and Individual
Constituents Commonly Display Hormesis Dose Responses, Especially for
Neuroprotective Effects. Molecules. 2020; 25 doi: 10.3390/molecules25112719.
21. Calabrese EJ. (2016). Preconditioning is hormesis part II: How the conditioning dose
mediates protection: Dose optimization within temporal and mechanistic
frameworks. Pharmacol.Res. 2016; 110: 265–275. doi: 10.1016/j.phrs.2015.12.020.
22. Calabrese V, Cornelius C, Calabrese EJ, Mattson MP (2010). Cellular stress
responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic
intervention in neurodegenerative disorders. Antioxid. Redox Signal. 13: 1763–
1811. doi: 10.1089/ars.2009.3074.
23. Caliskan B, Guven A, Ozler M, CAYCI T, OZCAN A, BEDIR O, SURER I,
KORKMAZ A. (2011). Ozone therapy prevents renal inflammation and fibrosis in a
rat model of acute pyelonephritis. Scand J Clin Lab Invest. 2011; 71: 473–480. doi:
10.3109 / 00365513.2011.587022.
24. Clavo B, Martínez-Sánchez G, Rodríguez-Esparragón F, Rodríguez-Abreu D, Galván
S; Aguiar-Bujanda D, Díaz-Garrido JA, Cañas S, Torres-Mata LB, Fabelo H; et al.
(2021). Modulation by Ozone Therapy of Oxidative Stress in Chemotherapy-Induced
Peripheral Neuropathy: The Background for a Randomized Clinical Trial. Int. J. Mol.
Sci. 22: 2802. https://doi.org/ 10.3390/ijms22062802.
25. Clavo B, Rodríguez-Esparragón F, Rodríguez-Abreu D, Martínez-Sánchez G, Llontop
P, Aguiar-Bujanda D, Leandro Fernández-Pérez L, a Santana-Rodríguez S (2019).
Modulation of Oxidative Stress by Ozone Therapy in the Prevention and Treatment of
Chemotherapy-Induced Toxicity: Review and Prospects. 8(12): 588.
26. Clavo , B , Suarez G, Aguilar Y, Gutierrez D, Ponce P, Cubero A, Robaina
F, Carreras JL (2011). Brain ischemia and hypometabolism treated by ozone therapy.
[PubMed]. 18(5): 283-7. doi: 10.1159/000333795.
27. Clavo B, Juan L Pérez L, López L, Suárez G , Lloret M, Rodríguez V , Macías
D , Santana M , Hernández MA , Oliva RM , Robaina F (2004). Ozone Therapy for
Tumor Oxygenation: a Pilot Study. [PubMed]. Evid Based Complement Alternat
Med. 1(1): 93-98. doi: 10.1093/ecam/neh009.
28. Currò M, Russo T, Ferlazzo N, Caccamo D, Antonuccio P, Arena S, Parisi S, Perrone
P, Ientile R, Romeo C, Impellizzeri P (2018). Anti-Inflammatory and Tissue
Regenerative Effects of Topical Treatment with Ozonated Olive Oil/Vitamin E
Acetate in Balanitis Xerotica Obliterans. Molecules. 23(3): 645. doi:
10.3390/molecules23030645. PMID: 29534008; PMCID: PMC6017296.
29. Dahhan M KB (2015). Cases Study of the Effectiveness of Ozone Gas Therapy on the
Treatment of Resistant Meningitis in Infants with Hydrocephalus. J Neurol Stroke
3(5): 00107. doi: 10.15406/jnsk.2015.03.00107.
30. Dattilo S, Mancuso C, Koverech G, Di Mauro P, Ontario ML, Petralia CC, Petralia A,
Maiolino L, Serra A, Calabrese EJ, Calabrese V (2015). Heat shock proteins and
hormesis in the diagnosis and treatment of neurodegenerative diseases. Immun
Ageing. 12: 20. doi: 10.1186/s12979-015-0046-8. PMID: 26543490; PMCID:
PMC4634585.
15
31. De Oliveira Magalhaes FN, Dotta L, Sasse A, MJ, Erich T Fonoff. (2012). Ozone
therapy as a treatment for low back pain secondary to herniated disc: a systematic
review and meta-analysis of randomized controlled trials. Pain Physician. 15(2):
E115-29.
32. Delgado-Roche L, Riera-Romo M, Mesta F, Hernandez-Matos Y, Barrios JM,
Martinez-Sanchez G, Al-Dalaien SM. (2017). Medical ozone promotes Nrf2
phosphorylation reducing oxidative stress and pro-inflammatory cytokines in multiple
sclerosis patients. Eur. J. Pharmacol. 811: 148–154. doi:
10.1016/j.ejphar.2017.06.017.
33. Delgago-Roche L, Riera-Romo M, Mesta F, Hernández-Matos Y, Juan M. Barrios JM,
Martínez-Sánchez G,Al-Dalaien SM(2017). Medical ozone promotes Nrf2
phosphorylation reducing oxidative stress and pro-inflammatory cytokines in multiple
sclerosis patients. [PubMed]. 2017.15;811:148-154. doi:
10.1016/j.ejphar.2017.06.017.
34. Dugger BN, Dickson DW (2017). Pathology of Neurodegenerative Diseases. Cold
Spring Harb Perspect Biol. 9(7): a028035. doi: 10.1101/cshperspect.a028035.
35. Elvis AM, Ekta JS (2011). Ozone therapy: A clinical review. J Nat Sci Biol Med. 2(1):
66-70. doi: 10.4103/0976-9668.82319.
36. Fernández-Leon OS, Ajamieh HH, Berlanga J, et al. (2008). Ozone oxidative
preconditioning is mediated by A1 adenosine receptors in a rat model of liver
ischemia/reperfusion. Transpl Int. 21: 39–48. doi: 10.1111 / j.1432-
2277.2007.00568.x
37. Galie M, Costanzo M, Nodari A, Boschi F, Calderan L, Mannucci S, Covi V.,
Tabaracci G., Malatesta M. (2018). Mild ozonisation activates antioxidant cell
response by the Keap1/Nrf2 dependent pathway. Free Radic. Biol. Med. 124: 114–
121. doi: 10.1016/j.freeradbiomed.2018.05.093
38. Gao L, Chen R-W, Williams JP, Li T, Han W-J, Qian-Nan Zhao Q-N, Yong Wang Y,
An J-X (2020). Efficacy and Safety of Percutaneous Ozone Injection Around
Gasserian Ganglion for the Treatment of Trigeminal Neuralgia: A Multicenter
Retrospective Study. Pain Res. 13: 927–936. Published online 2020 May 4.
doi: 10.2147/JPR.S232081.
39. García-Escudero V, Martín-Maestro P, Perry G, Avila J (2013). Deconstructing
mitochondrial dysfunction in Alzheimer disease. Oxid Med Cell Longev. 2013:
162152. doi: 10.1155/2013/162152.
40. González R, Borrego A, Zamora Z, Romay C, Hernández F, Menéndez S, Montero T,
Rojas E (2004). Reversion by ozone treatment of acute nephrotoxicity induced by
cisplatin in rats. Mediators Inflamm. 13(5-6): 307-312. doi:
10.1080/09629350400008836.
41. Gulmen S, Kurtoglu T, Meteoglu I, Kaya S, Okutan H. (2013). Ozone therapy as an
adjunct to vancomycin enhances bacterial elimination in methicillin resistant
Staphylococcus aureus mediastinitis. J Surg Res. 185: 64–69. doi: 10.1016 /
j.jss.2013.05.085.
42. Guven A, Gundogdu G, Sadir S, Topal T, Erdogan E, Korkmaz A, Surer I, Ozturk H.
(2008). The efficacy of ozone therapy in experimental caustic esophageal burn. J.
Pediatr. Surg. 43: 1679–1684. doi: 10.1016/j.jpedsurg.2008.01.064.
43. Higdon AN, Landar A, Barnes S, Darley-Usmar VM (2012). The electrophile
responsive proteome: integrating proteomics and lipidomics with cellular function.
Antioxid Redox Signal. 17(11): 1580-9. doi: 10.1089/ars.2012.4523.
16
44. Hohn A, Tramutola A, Cascella R. (2020). Proteostasis Failure in Neurodegenerative
Diseases: Focus on Oxidative Stress. Oxid Med Cell Longev. 2020 (Special issue):
Article ID 5497046. doi.org/10.1155/2020/5497046
45. Hu J, Ma H, Zhu S, Cai X, Yan K, Lu HD. ( 2018). Visual Motion Processing in
Macaque V2. Rec. Oct. 2016, 2018. Publ.: Oct.2018.
doi.org/10.1016/j.celrep.2018.09.014.
46. Chang JD, Lu HS, Chang YF, Wang D. (2005). Ameliorative effect of ozone on
cytokine production in mice injected with human rheumatoid arthritis synovial
fibroblast cells. Rheumatol Int. 26: 142–151. doi: 10.1007/s00296-004-0526-1.
47. Chang ET, SmedbyShumin KE, Zhang M, Hjalgrim H, Melbye M, Ost A, Glimelius
B, Wolk A, Adami HO. (2005). Dietary factors and risk of non-hodgkin lymphoma in
men and women. Cancer Epidemiol Biomarkers Prev . 2005 Feb;14(2):512-20. doi:
10.1158/1055-9965.EPI-04-0451.
48. Inal M, Dokumacioglu A, Ozcelik E, Ucar O (2011).The effects of ozone therapy and
coenzyme Q₁₀ combination on oxidative stress markers in healthy subjects. Ir J Med
Sci. 180(3): 703-7. doi: 10.1007/s11845-011-0675-7.
49. Ishii T, Itoh K, Ruiz E, Leake DS, Unoki H, Yamamoto M, Mann GE. (2004). Role of
Nrf2 in the regulation of CD36 and stress protein expression in murine macrophages:
activation by oxidatively modified LDL and 4-hydroxynonenal. Circ. Res. 94 (5):
609–616. https://doi.org/10.1161/01.RES.0000119171.44657.45
50. Izadi M, Kheirjou R, Mohammadpour R, Aliyoldashi MH, Moghadam SJ, Khorvash
F, Jafari NJ, Shirvani S, Khalili N. (2019). Efficacy of comprehensive ozone therapy
in diabetic foot ulcer healing. Diabetes Metab Syndr. 13(1): 822–825. doi:
10.1016/j.dsx.2018.11.060.
51. Kakkad VJ (2018). Neurological deficits ltreated with Ozone Therapy. Proceedings of
the 5Th WFOT Meeting; 2016 Nov 18-20; Mumbai, India. J Ozone Therapy. 2(2).
doi:10.7203/jo3t.2.2.2018.11155.
52. Kal1 E, ProseÂe1R, Winters M, van der Kamp J, (2017). Does implicit motor learning
lead to greater automatization of motor skills compared to explicit motor learning? A
systematic review. Systematic review. Rec.: October 31, 2017, Published: September
5, 2018, PLoS ONE 13(9): Journal.pone.0203591 doi.org/10.1371/.
53. Knowles TP, Vendruscolo M, Dobson CM. (2014). The amyloid state and its
association with protein misfolding diseases. Nat Rev Mol Cell Biol. 15 (6): 384–396.
doi: 10.1038/nrm3810.
54. Lahodny J. (2021). Ozonanwendungen [online]. 2021. cit. 2021-02-03). Available on
the Internet: https://dr-lahodny.at/ozonanwendungen/
55. Leri M, Scuto M, Ontario ML, Calabrese V, Calabrese EJ, Bucciantini M, Stefani M
(2020). Healthy Effects of Plant Polyphenols: Molecular Mechanisms. Int J Mol Sci.
21(4): 1250. doi: 10.3390/ijms21041250.
56. Levonen AL, Landar A, Ramachandran A, Ceaser EK, Dickinson DA, Zanoni G,
Morrow JD, Darley-Usmar VM. (2004). Cellular mechanisms of redox cell signalling:
role of cysteine modification in controlling antioxidant defences in response to
electrophilic lipid oxidation products. Biochem. J. 378 (Pt2): 373–382. Doi:
10.1042/BJ20031049.
57. Li J, Li T, Li G, Liu H, Zhang X. (2020). Selective nerve root injection of ozone for
the treatment of phantom limb pain: Three case reports. 99(16): e19819. doi:
10.1097/MD.000000000000019819.
58. Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G,
Cacciatore F, Bonaduce D, Abete P. (2018). Oxidative stress, aging, and diseases.
17
Clin. Interv. Aging 13, 757–772. Clin Interv Aging. 13: 757–772.
doi: 10.2147/CIA.S158513.
59. Madej P, Plewka A, Madej JA, et al. (2007). Ozonotherapy in an induced septic
shock. I. Effect of ozonotherapy on rat organs in evaluation of free radical reactions
and selected enzymatic systems. Inflammation. 30: 52–58. doi: 10,1007 / s10753-
007-9021-7.
60. Mancuso C, Capone C, Ranieri SC, et al (2008). Bilirubin as an endogenous
modulator of neurotrophin redox signaling. J Neurosci Res. (10)86: 2235–2249.
61. Mancuso C, Pistritto G, Tringali G, Grossman AB, Preziosi P, Navarra P (1997).
Evidence that carbon monoxide stimulates prostaglandin endoperoxide synthase
activity in rat hypothalamic explants and in primary cultures of rat hypothalamic
astrocytes. Brain Res Mol Brain Res.PubMed. 45(2): 294-300. doi: 10.1016/s0169-
328x (96)00258-6.
62. Mancuso C. (2017). Bilirubin and brain: a pharmacological
approach. Neuropharmacology. 118:113–123. doi: 10.1016 /
j.neuropharm.2017.03.013.
63. Manoto SL, Maepa MJ, Motaung SK. (2018). Medical ozone therapy as a potential
treatment modality for regeneration of damaged articular cartilage in osteoarthritis.
Saudi J Biol Sci. 25: 672–679. doi: 10.1016/j.sjbs.2016.02.002
64. Massaad CA (2011). Neuronal and vascular oxidative stress in Alzheimer's disease
Curr Neuropharmacol. PubMed. 2011 Dec;9(4):662-73. doi:
10.2174/157015911798376244.
65. Mašán J, Golská S. (2017). Infratentorial ependymoma of a two-year-old child,
neurorehabilitation and ozone therapy. In: Neurorehab 2017. print.: ALMIL, p. 34-35.
ISBN 978-80-971938-4-3.
66. Mašán J. (2017). Use ozone in neurology. In: Nerorehab 2017. print. Almil.
Page:120-126. ISBN 978-80-85659-84-9.
67. Mašán J. (2018). Ozone therapy in traumatology. In: Pitfalls and complications in the
treatment of fractures . print.: Galén. p. 396-400. 2018. ISBN 978-80-7492-393-7.
68. Mitrečić D , Petrović DJ, Stančin P, Isaković J, Zavan B, Tricarico G, Kujundžić
Tiljak M, Di Luca M. (2020). How to face the aging world - lessons from dementia
research . Croat Med J. 61(2): 139-146. doi: 10.3325/cmj.2020.61.139.
69. Molinari F, Rimini D, Liboni W, et al. ( 2017). Cerebrovascular pattern improved by
ozone autohemotherapy: an entropy-based study on multiple sclerosis patients. Med
Biol Eng Comput. 55: 1163–1175. doi: 10,1007 / s11517-016-1580-z.
70. Molinari F, Simonetti V, Franzini M, Pandolfi S, Vaiano F, Valdenassi L, Liboni W
(2014). Ozone autohemotherapy induces long-term cerebral metabolic changes in
multiple sclerosis patients . 27(3): 379-89. doi: 10.1177/039463201402700308.
71. Pedruzzi LM, Stockler-Pinto MB, Leite JrM, Mafra D. (2012). Nrf2-keap1 system
versus NF-kappaB: the good and the evil in chronic kidney disease? Biochimie. 94
(12): 2461-2466. doi: 10.1016/j.biochi.2012.07.015
72. Pryor R, Norvaisas P, Marinos G, Best L, Thingholm LB, Quintaneiro LM, De Haes
W, Esser D, Waschina S, Lujan C, Smith RL, Scott TA, Martinez-Martinez D,
Woodward O, Bryson K, Laudes M, Lieb W, Houtkooper RH, Franke A, Temmerman
L, Bjedov I, Cocheme HM, Kaleta C, & Cabreiro F (2019). Host-Microbe-Drug-
Nutrient Screen Identifies Bacterial Effectors of Metformin Therapy. Cell.
doi:10.1016/j.cell.2019.08.003.
73. Qiu A, Zhang H, Wang Ch, Chong YS, Shek LP, Gluckman PD, Meaney MJ, Fortier
MV & Wu Y. (2021). Canonical TGF-β signaling regulates the relationship between
18
prenatal maternal depression and amygdala development in early life. Translational
Psychiatry, Volume 11, Article Number: 170 (2021). Published: 15 March 2021.
74. Qiu J, Chen HS (2016). Efficacy and safety of ozone therapy administered by
autologous blood transfusion for acute ischemic stroke: study protocol for a multi-
center open-label large-sample parallel randomized controlled trial. Asia Pac. Journal:
Nerv Syst Dis. IP: 213.215.84.6]. 1(2): 37-42. doi: 10.4103/2468-5577.181233.
75. Ramirez-Acuña JM, Cardenas-Cadena SA, Marquez-Salas PA, Garza-Veloz I, Perez-
Favila A, Cid-Baez MA, Flores-Morales V, Martinez-Fierro ML (2019). Diabetic Foot
Ulcers: Current Advances in Antimicrobial Therapies and Emerging Treatments.
Antibiotics (Basel). 8(4): 193. doi: 10.3390/antibiotics8040193.
76. Rattan S, Demirovic D (2009). Hormesis can and does work in humans. [PubMed].
Dose Response.8(1): 58-63. doi: 10.2203/dose-response.09-041.
77. Re L, Martinez-Sanchez G., Bordicchia M, Malcangi G, Pocognoli A, Morales-Segura
MA, Rothchild J, Rojas A (2014). Is ozone pre-conditioning effect linked to
Nrf2/EpRE activation pathway in vivo? A preliminary result. Eur. J. Pharmacol. 742:
158–162. doi: 10.1016/j.ejphar.2014.08.029.
78. Reutzel M, Grewal R, Dilberger B, Silaidos C, Joppe A, Eckert GP (2020). Cerebral
Mitochondrial Function and Cognitive Performance during Aging: A Longitudinal
Study in NMRI Mice. Oxid Med Cell Longev. 2020:
4060769. doi: 10.1155/2020/4060769.
79. Rowen RJ (2018). Ozone therapy in conjunction with oral antibiotics as a successful
primary and sole treatment for chronic septic prosthetic joint: review and case report.
Med Gas Res. 8(2): 67-71. doi: 10.4103/2045-9912.235139.
80. Scassellati C, Ciani M, Galoforo A.C, Zanardini R, Bonvicini C, Geroldi C (2020).
Molecular mechanisms in cognitive frailty: potential therapeutic targets for oxygen-
ozone treatment. Mech. Ageing Dev. 186: 111210. doi: 10.1016/j.mad.2020.111210.
81. Scassellati C, Costanzo M, Cisterna B, Nodari A, Galie M, Cattaneo A, Covi
V,Tabaracci G, Bonvicini C, Malatesta M. (2017). Effects of mild ozonisation on gene
expression and nuclear domains organization in vitro. Toxicol In Vitro. 44: 100-110.
doi: 10.1016/j.tiv.2017.06.021.
82. Scassellati C, Galoforo AC, Bonvicini C , Esposito C , Ricevuti G (2020). Ozone: a
natural bioactive molecule with antioxidant property as potential new strategy in aging
and in neurodegenerative disorders. Ageing Res Rev. 63:101138. doi:
10.1016/j.arr.2020.101138.
83. Schmidlin CJ, Dodson MB, Madhavan L, Zhang DD (2019). Redox regulation by
NRF2 in aging and disease. Free Radic. Biol. Med. 134: 702–707. doi: 10.1016 /
j.freeradbiomed.2019.01.016.
84. Silva-Palacios A, Ostolga-Chavarria M, Zazueta C, Konigsberg M. (2018). Nrf2:
Molecular and epigenetic regulation during aging. Ageing Res. Rev. 47:31–40. doi:
10.1016/j.arr.2018.06.003.
85. Simonetti, V, Liboni W, Molinari F (2014). Why Ozone Therapy in Multiple
Sclerosis?. Revista Española de Ozonoterapia. 4(1): 51-68. ISSN: 2174-3215
86. Singh A, Kukreti R, Saso L, Kukreti S (2019). Oxidative Stress: A Key Modulator in
Neurodegenerative Diseases. Molecules. 24 doi: 10.3390/molecules24081583.
87. Siniscalco D, Trotta MC, Brigida AL, Maisto R, Luongo M, Ferraraccio F, D'Amico
M, Di Filippo C (2018). Intraperitoneal Administration of Oxygen/Ozone to Rats
Reduces the Pancreatic Damage Induced by Streptozotocin. Biology (Basel) 7(1): 10.
doi: 10.3390/biology7010010.
19
88. Smith NL, Wilson AL, Gandhi J, Vatsi S, Khan SA (2017). Ozone therapy: an
overview of pharmacodynamics, current research, and clinical utility. Med. Gas Res.
7(3): 212-219. doi: 10.4103/2045-9912.215752.
89. Tang, T, D. Shindell, G. Faluvegi, G. Myhre, D. Olivié, A. Voulgarakis, M. Kasoar, T.
Andrews, O. Boucher, P.M. Forster, Ø. Hodnebrog, T. Iversen, A. Kirkevåg, J.-F.
Lamarque, T. Richardson, B.H. Samset, C.W. Stjern, T. Takemura, and C. Smith.
(2019). Comparison of effective radiative forcing calculations using multiple methods,
drivers, and models. J. Geophys. Res. Atmos., 124, no. 8, 4382-4394,
doi:10.1029/2018JD030188.
90. Tirelli U, Cirrito C, Pavanello M, Piasentin C, Lleshi A, Taibi R. (2019). Ozone
therapy in 65 patients with fibromyalgia: an effective therapy. Eur. Rev. Med.
Pharmacol. Sci. 23:1786–1788.
91. Toda N, Ayajiki K, Okamura T. (2009). Cerebral blood flow regulation by nitric
oxide: recent advances Pharmacol Rev. PubMed 2009 Mar;61(1):62-97. doi:
10.1124/pr.108.000547.
92. Vaillant JD, Fraga A, Diaz MT, Mallok A, Viebahn-Hansler R, Fahmy Z, Barbera A,
Delgado L, Menendez S, Fernandez OS. (2013). Ozone oxidative postconditioning
ameliorates joint damage and decreases pro-inflammatory cytokine levels and
oxidative stress in PG/PS-induced arthritis in rats. Eur. J. Pharmacol. 714: 318–324.
doi: 10.1016/j.ejphar.2013.07.034
93. Valacchi G, Bocci V (2000). Studies on the biological effects of ozone: 11. Release of
factors from human endothelial cells. Mediators Inflamm. 9:271–276. doi:
10.1080/09629350020027573.
94. Wang L, Chen Z, Liu Y, Du Y, Liu X. (2018). Ozone oxidative postconditioning
inhibits oxidative stress and apoptosis in renal ischemia and reperfusion injury through
inhibition of MAPK signaling pathway. Drug Des Devel Ther. 12: 1293–1301.
Doi:147/DDDT.S164927.
95. Yanar K, Atayik MC, Simsek B, Çakatay U (2020). Novel biomarkers for the
evaluation of aging-induced proteinopathies. Biogerontology. 21(5): 531-548. doi:
10.1007/s10522-020-09878-8.
96. Yuldashev SS, Mamadaliev AM, Aliev MA (2020). Endolumbal Nootropic-Ozone
Therapy In Complex Treatment Of Patients With Complicated Spinal Injury In Acute
Period. 2020. Shavkiddin S. Yuldashev Abdurakhmon M. Mamadaliev Mansur A.
Aliev Eur J Molec Clin Med. 7(3): 1571-1576. Online ISSN: 2515-8260.
97. Yuldashev TK, Karimov ET. (2020). Inverse Problem for a Mixed Type Integro-
Differential Equation with Fractional Order Caputo Operators and Spectral
Parameters. Journals Axioms, 2020. Volume 9, Issue 4, doi: 10.3390/axioms9040121
98. Zhang J, Davies KJA, Forman H.J (2015). Oxidative stress response and Nrf2
signaling in aging. Radic Biol Med. 88:314–336. doi:
10.1016/j.freeradbiomed.2015.05.036.
99. Zhang J, Guan M, Xie C, Luo X, Zhang Q, Xue Y (2014). Increased growth factors
play a role in wound healing promoted by noninvasive oxygen-ozone therapy in
diabetic patients with foot ulcers. Oxid Med Cell Longev. 2014: 273475. doi:
10.1155/2014/273475.
100. Zhao X, Li Y, Lin X, Wang J, Zhao X, Xie J, Sun T, Fu Z. (2018). Ozone
induces autophagy in rat chondrocytes stimulated with IL-1beta through the
AMPK/mTOR signaling pathway. J Pain Res. 2018 (11): 3003–3017.
doi.org/10.2147/JPR.S183594
20
101. Zhou M, Hou J, Li Y, Mou S, Wang Z, Horch RE, Sun J, Yuan Q (2019). The
pro-angiogenic role of hypoxia inducible factor stabilizer FG-4592 and its application
in an in vivo tissue engineering chamber model. Sci. Rep. 9(1): 6035. doi:
10.1038/s41598-019-41924-5.