Next Article in Journal
Oral Health-Related Quality of Life in Native and Immigrant Populations in the PELFI Study in Spain
Previous Article in Journal
Removal of Sulfamethoxazole, Sulfathiazole and Sulfamethazine in their Mixed Solution by UV/H2O2 Process
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Can Hypoxic Conditioning Improve Bone Metabolism? A Systematic Review

by
Marta Camacho-Cardenosa
1,*,
Alba Camacho-Cardenosa
1,
Rafael Timón
1,
Guillermo Olcina
1,
Pablo Tomas-Carus
2,3 and
Javier Brazo-Sayavera
4,5
1
Faculty of Sport Science, University of Extremadura, 10003 Cáceres, Spain
2
Departamento de Desporto e Saúde, Escola de Ciência e Tecnologia, Universidade de Évora, 7000-812 Évora, Portugal
3
Comprehensive Health Research Centre (CHRC), University of Évora, 7000-812 Évora, Portugal
4
Instituto Superior de Educación Física, Universidad de la República, 40000 Rivera, Uruguay
5
Polo de Desarrollo Universitario EFISAL, Universidad de la República, 40000 Rivera, Uruguay
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2019, 16(10), 1799; https://doi.org/10.3390/ijerph16101799
Submission received: 16 April 2019 / Revised: 14 May 2019 / Accepted: 15 May 2019 / Published: 21 May 2019
(This article belongs to the Section Health Behavior, Chronic Disease and Health Promotion)

Abstract

:
Among other functions, hypoxia-inducible factor plays a critical role in bone–vascular coupling and bone formation. Studies have suggested that hypoxic conditioning could be a potential nonpharmacological strategy for treating skeletal diseases. However, there is no clear consensus regarding the bone metabolism response to hypoxia. Therefore, this review aims to examine the impact of different modes of hypoxia conditioning on bone metabolism. The PubMed and Web of Science databases were searched for experimental studies written in English that investigated the effects of modification of ambient oxygen on bone remodelling parameters of healthy organisms. Thirty-nine studies analysed the effect of sustained or cyclic hypoxia exposure on genetic and protein expression and mineralisation capacity of different cell models; three studies carried out in animal models implemented sustained or cyclic hypoxia; ten studies examined the effect of sustained, intermittent or cyclic hypoxia on bone health and hormonal responses in humans. Different modes of hypoxic conditioning may have different impacts on bone metabolism both in vivo and in vitro. Additional research is necessary to establish the optimal cyclical dose of oxygen concentration and exposure time.

1. Introduction

Our understanding of the influence of hypoxia on human physiology has been improved with the discovery of hypoxia-inducible factor (HIF). Low available oxygen leads to the decline of oxygen partial pressure in arterial blood (PiO2), which determines HIF stabilisation [1]. HIF translocates to the nucleus where it increases the mRNA expression of a wide variety of genes [2]. Among others, respiratory rate, heart rate and systemic blood pressure are increased to improve oxygen transport and utilisation [3]. If the exposure is maintained for a long period of time, a switch from mitochondrial oxidative phosphorylation to anaerobic glycolysis occurs with concomitant production of reactive oxygen species (ROS) [4]. Whereas higher levels of ROS could be detrimental [5], low levels of ROS can have protective effects on specific cells, tissues and organs [6].
Although hypoxia-driven pathways are vital in normal development and tissue homeostasis, potential mechanisms resulting in osteoblastic activity remain unclear [7]. HIF mediates upregulation of glycolytic enzymes such as pyruvate dehydrogenase kinase 1 (PDK1), lactate dehydrogenase A (LDHA) and glucose transporters (GLUTs) that compensate for the energy inefficiency of glycolysis [8]—the main metabolic pathway during osteoblastic differentiation [9]. Even in the presence of oxygen, bone cells metabolise glucose by a phenomenon known as the “Warburg effect” or “aerobic glycolysis” [10]. Thus, it could be hypothesised that hypoxic exposure could enhance bone formation by promoting glycolysis as the main metabolic pathway. However, HIF also regulates bone remodelling-related genes, such as vascular endothelial growth factor (VEGF), erythropoietin (EPO) and osteoprotegerin (OPG)—the factor that inhibits osteoclastogenesis by counteracting RANKL and therefore, bone reabsorption [11]. Because of the vital role in angiogenic–osteogenic coupling, animal and cell studies have suggested that hypoxic conditioning (HC) could be a potential nonpharmacological strategy for treating hypoxic-ischaemic diseases, including skeletal diseases [12].
HC is a drug-free method based on the adaptation produced by breathing air with low oxygen content [4]. During the last 30 years, HC interventions have enhanced physical and mental functions and the prevention of aging and different diseases in two million patients [13]. HC can be implemented with different patterns and severity, mediating different molecular pathways [14]. An optimal dose in terms of duration, frequency and severity could range from no response at low intensity to a protected state and higher intensities or even a further increase in stimulus could cause negative adaptations [15]. In this sense, some authors have distinguished between different hypoxia modes. “Sustained hypoxia” is characterised by a single episode of hypoxia, which is maintained during a prolonged stay. Hypoxia can also be interrupted by reoxygenation periods and, thus, two modes of hypoxia should be differentiated. “Intermittent hypoxia” is characteristic of obstructive sleep apnoea syndrome (OSAS), where HIF-1 activation is stimulated by many short cycles of severe hypoxia lasting 15–30 s and longer periods of reoxygenation. However, during “cyclical hypoxia”, HIFs accumulate strictly during longer periods of moderate hypoxia (since 12 h), followed by identical reoxygenation periods [14]. Previous reviews have considered the effects of sustained, intermittent and cyclical exposures on cardiovascular and respiratory physiology, health and overall quality of life [1,4,7,13]. It has been shown that “sustained” and “intermittent” hypoxia could lead to deleterious consequences by increasing oxidative stress [16] and producing systemic inflammation [17], whereas “cyclical” hypoxia may lead to a prolonged and sustained state of protection [15]. However, there is currently not a clear consensus regarding the response of bone metabolism to hypoxia [18] and therefore, the optimal dose for HC to achieve beneficial effects is unknown. Due to the lack of systematic reviews on this topic, it will be important to merge the information with the available evidence. Thus, the aim of the present review was to examine the impact of the different modes of HC on bone metabolism.

2. Materials and Methods

2.1. Searches and Article Selection Strategy

This systematic review was carried following Preferred Reporting Item for Systematic Reviews and Meta-Analyses (PRISMA) methodology [19]. The well-known electronic databases selected were Web of Science and PubMed. The selected articles were original articles published between 1900 and 2019. Search terms used: “hypoxi*”, “altitude”, “oxygen deprivation”, ”bone remodelling”, “bone metabolism”, “osteog*”, “bone tissue remodelling”, “bone mineral density”, “growth*”, “pulmonary”, “cancer” and “tumour”. The exact search strategy can be found in Appendix A. The search was finalised on the 1 March 2019. The main author deleted duplicate articles.
Inclusion criteria that were defined for article selection: (1) written in English; (2) experimental study on bone remodelling parameters; (3) containing a sample of healthy organisms; and (4) application of a hypoxic treatment (modification of ambient oxygen). Studies were excluded if they were only presented once as a conference, congress or seminar. These criteria were evaluated first by the titles and abstracts by two authors (A.C.-C. and M.C.-C.) independently. If they met the inclusion criteria or if the title and abstract did not provide sufficient information, full articles of these studies were obtained to apply the criteria at full text by the same two authors. To resolve any data discrepancies, group discussions were conducted until a mutual consensus was reached. A third expert (J.B.-S.) was consulted when a consensus could not be reached.

2.2. Risk of Bias

The internal quality of each study was assessed using the Office of Health Assessment and Translation (OHAT) Risk of Bias Rating Tool [20]. Two review authors (M.C.-C. and A.C.-C.) independently assessed 11 risk of bias questions using the 4-point scale ranging from low to high risk of bias options. Discrepancies between reviewers were resolved using consensus. For both in vivo and in vitro studies, an initial confidence rating was given, which was subsequently downgraded or upgraded according to factors that decrease or increase confidence in the results [21,22].
To establish a credible link between hypoxic exposure and bone health effect, confidence ratings were assigned to individual study designs and translated into a level of evidence (“high”, “moderate”, “low”, “evidence of no health effect” and “inadequate evidence”).

2.3. Data Extraction

One author (M.C.-C.) applied the data extraction. Another author (J.B.-S.) verified this extraction. Details were extracted regarding (a) sample: type (i.e., human, animal or cell), age and sample size; (b) study design: conditions, exposure type or timing (i.e., normobaric or hypobaric; expansion, proliferation or differentiation); protocol (i.e., sustained, cyclic or intermittent) and duration, frequency and hypoxic level; and (c) effects of hypoxia on bone outcomes.

3. Results

3.1. Article Selection

The initial electronic database search resulted in a total of 39 citations in Web of Science and 236 in PubMed. After deleting 22 duplicates, 253 studies were analysed by title and abstract by applying the inclusion and exclusion criteria. A total of 167 studies were excluded and 86 potentially relevant studies were selected for full-text review. Fifty-two studies were identified in our systematic search from which risk of bias was described and data extracted. A flowchart of the search procedure can be found in Figure 1.

3.2. Risk of Bias

Results from the risk of bias assessment are shown in Appendix B and Appendix C. Evidence summarised from animal, human and in vitro studies suggested a range of likely high to definitely low level of confidence (see Appendix D). Selective reporting was considered the most pertinent domain, rating from likely low and definitely low for animal and human in vitro studies. Conversely, performance domain was identified at likely high or low level of confidence. This domain was likely high for all human studies.
In the association between hypoxic exposure and bone health effect (see Appendix E), moderate confidence ratings were assigned to epidemiological studies (human cohort and cross-sectional) whereas high confidence was established in experimental studies (in vitro, animal or human).

3.3. Data Extraction

Table 1 and Table 2 summarise the data from 39 in vitro and 13 in vivo studies. Table 1 shows the results of the extraction data of the in vitro studies. Experimental details from the in vivo studies can be found in Table 2.

3.3.1. In Vitro Studies

Thirty-nine studies analysed the effect of sustained or cyclic hypoxia exposure on bone remodelling parameters in different cell models from animals (14 studies) or humans (34 studies). Most commonly used stem cells models were: bone marrow stromal cells (BMSCs; n = 26), adipose-derived stromal cells (ASCs; n = 8), placental mesenchymal stem cells (pMSCs; n = 1), tendon-derived stem cells (TDSCs; n = 1), umbilical cord perivascular cells (UCPVCs; n = 1) and cartilage endplate stem cells (CESCs; n = 1). Only three of the in vitro studies reviewed exposed osteoblasts or osteocytes to sustained hypoxia [30,47,53]. Thirty-two studies included in this review applied sustained hypoxia protocols. Sustained hypoxia was administered from one to 28 days throughout the different timelines of cellular differentiation (expansion, n = 16; proliferation, n = 1; and differentiation, n = 27). Related to the hypoxia level, the dose ranged from 0.1 to 7% PiO2. Severe hypoxia (<3% PiO2) was applied more often (n = 33) than moderate hypoxia (>3% PiO2; n = 11). Only one study applied cyclic hypoxia (two bouts of 3 min per day) over the course of 15 days at 1, 3, 5 and 10% PiO2 in human BMSCs [58]. Runt-related transcription factor 2 (RUNX2; n = 15), alkaline phosphatase (ALP; n = 11), collagen type 1 alpha 1 (Col1A1; n = 6), osteopontin (OPN; n = 6) and osteocalcin (OC; n = 11) were the most studied genes. In terms of protein expression, RUNX2 (n = 5) and ALP (n = 14) were the predominant parameters. Mineralisation capacity was assessed using ALP activity (n = 17) and calcium deposits (n = 23).

3.3.2. In Vivo Studies

Three studies carried out in animal models implemented two different types of HC: sustained and cyclic hypoxia. All interventions involved daily exposure with a hypoxic level between 3000 and 6000 m over a prolonged period from 2 to 5 weeks [63,64,72]. Cyclic protocols adopted a pattern of exposure to hypoxia followed by the same time course of exposure to normoxia, lasting 4 [63] and 5 h [64] per day. Six out of the ten studies in humans, included in this review, analysed the effect of sustained or cyclic hypoxia in healthy active [60,61,62,65,66] or sedentary adults [67]. Three studies [60,61,62] implemented sustained HC corresponding to periods of 16, 24 and 60 weeks. The hypoxic level during these stays ranged from 2500 (moderate level) to 6700 m (severe level). Whereas Martinez-Guardado, (2019) and Ramos-Campos, (2015) [65,66] studied the effects after 7 and 8 weeks, respectively, of normobaric cyclic hypoxia training at 15% PiO2 (2 days per week; 60 min per session) and [67] applied bed rest or ambulatory normobaric hypoxia during 21 days at 4000 m of simulated altitude. Finally, four observational human studies analysed the effect of OSAS level (namely intermittent hypoxia) on bone remodelling parameters [68,69,70,71]. Healthy bone was evaluated with different parameters: bone volume (BV/TV; n = 2); trabecular number (Tb.N.; n = 2); bone mineral density-total (BMD-total; n = 6); bone mineral density-spine (BMD-spine; n = 3); speed of sound (SOS; n = 2) values of the radius, metatarsal and phalanx; and T-score of the radius and phalanx (n = 2). Additionally, bone formation and resorption markers as well as bone specific alkaline phosphatase (BAP; n = 2), ALP (n = 2), 25-hydroxy vitamin D3 (25-Vit D; n = 2), intact parathyroid hormone (i-PTH; n = 2), C-terminal propeptide of type I collagen (CICP; n = 2), N-telopeptide of type I collagen (NTX; n = 2), C-terminal telopeptide (CTX; n = 2), urinary DPD (n = 2), creatine ratio (DPD/Cr; n = 2) and OPG (n = 1) were analysed to determine the effects of different HC modes.

4. Discussion

The present review examined the impact of the different HC modes on bone metabolism. Overall, disparity in protocols, MSC sources and the composition of the cultive media [31,40] used has made it difficult to establish the role of HC in osteogenic differentiation [31]. Conversely, HC modes might have a different effect on skeletal health of animals and humans. A sustained hypoxic environment could negatively influence bone mass and bone quality when tissue PiO2 falls below 40 mmHg. However, short episodes with modest levels of hypoxia (9–16% PiO2) could lead to benefits [7] if it is administered repeatedly [73]. Finally, intermittent hypoxia (associated with OSAS) may have unfavourable effects on bone metabolism [69] and other organs and systems [74,75].

4.1. In Vitro Studies

4.1.1. Sustained Exposure

To evaluate the effects of hypoxia on osteogenic differentiation, the genetic and protein expression of different biomarkers were evaluated. Sustained hypoxia was administered in MSCs derived from bone marrow, adipose tissue, placental, tendon, umbilical cord or cartilage endplate from one to 28 days throughout the different timelines (expansion, proliferation or differentiation).
While some studies showed decreases in RUNX2 gene expression [23,27,42,44,45,47,49,50,53,57,72], expression was maintained in three studies [31,34,36]. A shorter exposure (3–14 days), applied in two studies, increased RUNX2 gene expression [32,34]. RUNX2 protein expression decreased in the five reviewed studies [27,39,48,49,53], which applied 5–21 days of sustained hypoxia at 1–2% PiO2. Overall, data from mRNA expression analysis could differ from protein expression by the post-translational modification of osteogenic biomarkers that may be a critical step dependent on the duration of hypoxia [26]. Osteogenic differentiation is controlled by RUNX2—a specific transcription factor that can promote or inhibit the expression of osteogenic differentiation-related genes [33]. Expression of RUNX2 can induce the synthesis of early (e.g., ALP and COL1A1) and late markers of osteoblast differentiation (e.g., OC) [31]. Thus, sustained hypoxic exposure of 1–5% PiO2 for 2–28 days show contradictory results and so how this exposition affects to the osteoblastic differentiation cannot establish.
Sustained hypoxia protocols of 1–2% PiO2 for 5–21 days showed lower ALP gene expression [27,53,72,76], while moderate hypoxia (2–5% PiO2) for 3–14 days showed greater expression [33,34,35]. Protein expression of ALP increased when lower severity (>2% PiO2) and length of exposure (2–21 days) were applied [26,32,34,55,76,77,78]. Finally, ALP activity decreased in 11 of 18 studies after severe hypoxia (1–2% PiO2) was applied for 2–28 days [23,31,42,45,47,49,52,53,54,57,77]. ALP is a biomarker of bone growth and development as it produces an alkaline environment allowing calcium to crystallise and strength to be achieved [27]. Thus, it seems that exposures with greater severity (1–2%) and longer duration (up to 28 days) could negatively affect ALP; however, protein expression increased when moderate hypoxia (<2%) was applied for a shorter period of time (up to 21 days).
Similar to other genes, Col1A1 expression showed contradictory results. While expression of the Col1A1 gene was maintained at 5% PiO2 [35,36], it decreased with 2% PiO2 for 7 [53] or 21 days [23], but increased with 2% PiO2 for 12 days [33]. Col1A1 is an indicator of the efficiency of the final osteogenic potential [79]. Therefore, a lower expression of COL1A1 would indicate a decreased efficiency in osteogenic potential [32]. In this sense, moderate oxygen concentration (2–5%) with a moderate exposure time may promote bone formation.
During bone formation, late in the mineralisation process, noncollagenous glycoproteins such as OPN and OC are abundant in the bone matrix with biological and mechanical functions of bone [80]. Decreases in OPN expression activate osteoclastic bone resorption and inhibit osteoblastic bone formation [81]. A lower genetic expression of OPN was observed following severe hypoxia protocols (1–2% PiO2) for 21 days [27,40,45,46,49], but OPN expression was maintained with 5% PiO2 during cellular expansion and differentiation of human ASCs [42]. Nine of the eleven studies that evaluated this parameter showed a lower expression of the OC gene when severe hypoxia protocols of 5–28 days were applied [27,40,45,46,49,72,76]. Only two studies [33,35] showed increased expression with a more moderate dose (>2%) and lower exposure time (3–12 days).
Finally, in the present review, calcium deposits maintained the same values compared with the normoxia groups in four studies, after low oxygen availability between 1% to 5% PiO2 was applied for 14–21 days [33,37,38,41]. In BMSCs, the most severe doses (<2%) showed decreased calcium deposits [27,29,40,41,46,49,51,52,76]. Previous researchers have suggested that there may exist a basal threshold of tissue oxygenation that regulates the deposition of minerals in the extracellular matrix [53]. Thus, similar to osteocytes buried in mineralised bone, lower PiO2 may result in low ALP activity and minimal mineralisation potential [53].
Overall, it is difficult to clarify the role of sustained HC on osteogenic differentiation [31]. The differences in reported effects on cellular behaviours may be due to disparity in protocols, MSC sources from different species or tissues and/or the composition of the cultive media [31,40].
Different effects have been reported due to discrepancies in oxygen concentration and exposure time. It is notable that 2% may represent a critical concentration, and therefore oxygen concentrations above 2% could promote osteogenic responses [31]. However, long-term or chronic exposure to hypoxia was reported to inhibit osteogenic differentiation. Effects of hypoxia on osteogenic differentiation may be time-dependent: osteogenesis could be accelerated in the early period, but sustained long-term hypoxia could result in poor osteogenesis [27]. On the other hand, previous studies have reported that under low O2 conditions, MSCs proliferate faster and for a longer period of time [29]. However, maintaining the exposure during cellular differentiation could maintain the undifferentiated characteristics of these cells [40]. Conversely, subsequent inductions under normoxic conditions during differentiation could maintain or improve the differentiation potential [82]. Thus, the timing of the exposure of MSCs to hypoxia could be important in osteogenic differentiation.
According to the origins of different tissues, MSCs show altered differentiation in response to hypoxia [48]. Osteogenesis may only be induced in periodontal ligament MSCs [83] under sustained hypoxic conditions but inhibited in bone marrow and adipose MSCs [50,57,84,85].

4.1.2. Cyclic Exposure

In a study conducted by [58], the effects of 15 days of different doses (1, 3, 5 and 10% PiO2) of cyclic hypoxia (two bouts of 3 min per day) during the differentiation of human BMSCs were studied. Compared with normoxia conditions, low oxygen concentration increased cell proliferation (especially at 3% PiO2) but inhibited osteoblastic differentiation by decreasing RUNX2 and OC gene expression. Thus, the exposure time used was not sufficient to promote osteogenic differentiation.

4.2. In Vivo Studies

4.2.1. Sustained Exposure

Some reports have indicated that residency at altitude may cause a marked deterioration in different indices of skeletal health [61]. The studies included in this review, which measured different indices of skeletal health in animals and humans, reported similar findings. At extreme altitude, healthy rats showed a decrement in the BV/TV, Tb.N. and BMD-total after 3 weeks of sustained simulated hypoxia at 6000 m [59]. In addition, 14 healthy young adults were exposed to bed rest or ambulatory normobaric hypoxia for 21 days at 4000 m of simulated altitude [67]. Bone mineral content-total (BMC-total) decreased after bed rest protocols and increased after ambulatory hypoxic conditions. In addition, at high altitude, a group of five healthy active male adults [60], who participated in an expedition of 24 weeks at 2500 m of altitude showed a decrease in BMD-spine. Also, the Indian army composed of healthy males stayed at high altitude (3450 m) for 16 weeks [62] and showed a decrease in bone strength (SOS values of the radius, metatarsal and phalanx, and T-score of the radius and phalanx). Thus, a sustained hypoxic environment could negatively influence the bone mass and bone quality when tissue PiO2 falls below 40 mmHg. HIF could affect the activity of multiple skeletogenic cells involved in angiogenesis, extracellular matrix formation and resistance to infection [59,86]. Nevertheless, hypoxic conditions could enhance the differentiation of osteoclasts [87] and modulate their binding to resorption sites [88].
In addition to skeletal health, prolonged residency in a hypoxic environment is associated with changes in turnover of bone metabolism coupled with specific endocrine adaptations. Bone formation markers such as ALP, bone-specific alkaline phosphatase (B-ALP), 25-Vit D, parathyroid hormone (PTH) and resorption markers (e.g., carboxy-terminal collagen cross-link (CTX), urinary DPD and creatine ratio (DPD/Cr)) were evaluated after a stay at extreme and high altitudes [61,62]. The study reported that after 4 months at extreme altitude, ALP, B-ALP and CTX decreased and the DPD/Cr ratio did not show any significant change. These results indicate activation of the bone resorption process at extreme altitude. The DPD/CR ratio, B-ALP, protein released by osteoblasts and CTX were lower at high altitude. Decreased formation and expression of bone resorption markers reflected a lower bone turnover at high altitude.
PTH is the major hormone regulating calcium metabolism [89]; this hormone aids in the production of bone-destroying osteoclasts and consequently speeds up bone remodelling and the release of Ca and other minerals in usable forms [90]. Whereas PTH levels are increased at extreme altitude and decreased at high altitude, serum 25-Vit D showed a significant decline at both high and extreme altitudes. Decline of 25-Vit D remains speculative but may be due to declined conversion of 25(OH) to 1.25(OH) D3 under conditions of low oxygen [61]. As a result, increased PTH may be required to increase this conversion to stimulate intestinal absorption of calcium. Calcium levels are maintained at extreme altitude and significantly increased at high altitude. These studies suggest that sustained hypoxia is associated with a decline in bone turnover due to reduced formation and expression of bone resorption markers. Whether this decline in bone turnover can lead to an increase in calcium deposition in bones during residency at high altitude remains to be determined [61]. At extreme altitudes, more significant changes occur in hormonal and biochemical bone remodelling parameters.
Compared to in vitro studies, in vivo environments are much more complex, and more factors related to the hypoxic environment may be responsible for the impaired bone strength and quality [59]. Weight loss [91], increased lean mass [65], lowered basal metabolic rate [59], decreased activity levels [92], insufficient vitamin D levels [60] or dietary changes in Ca2+ [93], vitamin C [94] or vitamin D [95] can also influence the BMD, and thus play an important role in healthy bone.

4.2.2. Cyclic Exposure

Different findings have been reported following the application of cyclic HC protocols in humans and animals. Increases in the BMD-spine of rats were observed after 5 weeks of cyclic normobaric hypoxia (5 days per week; 5 h per day) at 4500 m [64]. In addition, healthy active adults showed improved BMD-total after 8 weeks of normobaric hypoxic training [65]; however, in trained triathletes [66], 7 weeks of normobaric cyclic hypoxia training at 15% PiO2 (2 days per week; 60 min per session) resulted in no reported changes in BMD-total. In another study, cyclic HC exposure was applied for 2 weeks, 4 h per day at 3000 to 5000 m maintaining BV/TV, Tb.N., BMD-total and BMC-total level in healthy rats [63]. The variations in the present findings may be partly explained based on previous findings that explain how the numbers of hypoxic episodes, severity and duration of total exposure may result in different physiological responses [13]. In this sense, a small number of short episodes with modest levels of hypoxia (9–16% PiO2) could lead to benefits [7] administered repeatedly over days or weeks [73].
Arterial hypoxemia has been postulated to cause systemic inflammation by activation of regulatory pathways and cytokines, thus causing bone loss (see in vivo studies; Intermittent Exposure) [96]. However, rats exposed to cyclic normobaric hypoxia for 5 weeks showed higher BMD [64]. Increased ROS production will activate proinflammatory cytokines, which cause production of nitric oxide (NO) in osteoblasts and osteoclasts, among other cells. It is known that NO regulates osteoclast-mediated bone reabsorption, activating osteoblastic activity and inhibiting RANKL expression [8,97]. Inhibition of NO in these studies showed BMD levels significantly elevated as well, indicating that there are further mechanism(s) besides the NO-mediated effect in increasing BMD following cyclic hypoxic exposure, such as increased oxidative stress or a VEGF-mediated effect [64]. On the other hand, no significant changes in the level of Ca, P and PTH following hypoxic exposure could indicate a restrain of osteoclastic activity and/or stimulation of osteoblastic activity, affecting bone metabolism via multiple mechanisms. Overall, cyclic modes may inhibit osteoclastic activity and/or stimulate osteoblastic activity; more research is needed to understand these mechanisms [98].
Hormonal factors could also have an influence on the achieved effects. A group of ovariectomised rats were exposed to the same dose of hypoxia showing a decrease in the assessed outcomes [63]. This suggests that imbalanced bone remodelling caused by hypoxia occurs in female rats when oestrogen is deficient, leading to possible accelerated bone loss in postmenopausal women [63]. Although long-term exposure to cyclic moderate hypoxia could have benefits without detrimental effects, establishing the optimal cyclical dose in terms of episode duration and time of exposure for the treatment of skeletal diseases with low oxygen concentrations requires substantial additional research [73].

4.2.3. Intermittent Exposure

Nocturnal breathing difficulty, specially, sleep apnea, results in intermittently low oxygen levels by reductions of airflow while sleeping [99]. While some reports have studied the relationship between sleep apnea and bone health, it remains unclear. In the present review, observational human studies, which analyzed the effect of OSAS level on bone remodelling parameters [68], observed similar values in BMD-spine [70] and higher in BMD-total values compared with healthy adults. These findings could be explained by osteogenesis–angiogenesis coupling phenomenon, induced by HIF secretion [68]. HIF may promote osteogenic factors via VEGF or ALP expression [68]. However, gender, comorbidities, ethnic group or age could lead conflicting results. Usual comorbidities, characteristics of OSA patients, could affect to bone health and healthy controls could not be considered as a valid control. In this sense, others studies included in this review such as intermittent hypoxic expositions, which excluded OSA patients with comorbidities, showed lower BMD in femoral neck [69]. In addition, higher values of bone resorption markers (e.g., CTX) and similar OPG values were found in subjects with severe OSAS [69,71]. A previous review showed that OSAS’s patients had increased circulating markers of systemic inflammation, which may contribute to the development of osteopenia [96]. Chronic hypoxia could reduce the expression of bone formation markers such as B-ALP or type I collagen [88], and promote the function of osteoclasts by increasing cytokines such as interleukin-6 [88,100].
While some studies have shown that intermittent hypoxia may have a protective role in bone health [68], other studies with larger samples suggest that OSAS may have unfavourable effects on bone metabolism [69]. In addition, intermittent hypoxia, which is associated with OSAS, causes an increase in oxidative stress with negative effects on other organs and systems [74,75].

5. Conclusions

In conclusion, different modes of HC may lead to different impacts on bone metabolism in both in vivo and in vitro models. While sustained and intermittent hypoxia might inhibit osteogenic differentiation and promote osteoclast function, cyclical hypoxia has been presented as a promising strategy to beneficially impact bone metabolism. In this sense, moderate oxygen concentration (above 2% in vitro and 9–16% in vivo) administered repeatedly over days or weeks may promote mineralisation potential, inhibit osteoclast activity and/or stimulate osteoblast activity. However, additional research is necessary to establish the optimal cyclical dose in terms of oxygen concentration and exposure time (episode duration, number of exposures per day and length).

Author Contributions

M.C.-C., A.C.-C. and J.B.-S. revised the literature and evaluated risk bias; M.C.-C. and A.C.-C. drafted the manuscript; P.T.-C. and G.O. edited and revised the manuscript; J.B.-S. and R.T. approved the final version of the manuscript.

Funding

The project has been supported by the Government of Extremadura with funding from the European Regional Development Fund under Grant (Ref: GR18003); the European Fund for regional development (FEDER) allocated by European Union through the Horizon 2020 Program ALT20-03-0145-FEDER-000007 being part of a larger project entitled “Ageing Safely in Alentejo. Preventing Falls and Violence against the Elderly Understanding for Action—(ESACA)”; the Universidad de la República under Grant 003051-000603-16; and the Ministry of Education, Culture and Sports, under Grant FPU15/00450 and FPU15/00452.

Acknowledgments

We acknowledge the PDU EFISAL (Universidad de la República) for covering part of the costs of publishing this article in open access

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Appendix A.1. Search Strategies

PubMed:
((((((hypoxi*[Title] OR altitude[Title] OR “oxygen deprivation”[Title])) NOT growth*[Title]) NOT (tumour[Title/Abstract] OR cancer[Title/Abstract] OR pulmonary[Title/Abstract])) AND (“bone remodeling” OR “bone metabolism” OR osteog* OR “bone tissue remodeling” OR “bone mineral density”)) AND (“1900”[Date—Publication]: “3000”[Date—Publication])) AND English[Language]
Web of Science:
Title: (hypoxi* OR altitude OR “oxygen deprivation”) NOT Title:(growth*) NOT Topic: (tumour OR cancer OR pulmonary) AND Topic:(“bone remodeling” OR “bone metabolism” OR osteog* OR “bone tissue remodeling” OR “bone mineral density”)
Period of time: 1900–2019.
Language of search = English

Appendix B

Table A1. Risk of bias and confidence rating for in vitro studies.
Table A1. Risk of bias and confidence rating for in vitro studies.
ReferenceRisk of Bias QuestionsConfidence Rating
1234567891011
Bouvard, 2014--NANA+--+++++Moderate
Binder, 2015--NANA+-++++++Moderate
Burian, 2017--NANA+--++++Low
Ciappeti, 2016++-NANA+--+-+++High
Cicione, 2013--NANA+--+++++Moderate
D’Ippolito, 2006--NANA+--+++++Moderate
Deschepper, 2011--NANA----++++Moderate
Ding, 2014--NANA++--++++++High
Gao, 2013--NANA+--+++++Moderate
Gu, 2016--NANA---+++++Moderate
Holwarth, 2010--NANA+-+++++Low
Hopper, 2015+-NANA+---+++Low
Hsu, 2013--NANA----+++Low
Huang, 2011--NANA+---+++Low
Huang, 2012--NANA+--+++++Moderate
Iacono, 2018--NANA+--++++Low
Inagaki, 2017--NANA+++-+++++High
Jin, 2010--NANA+--+-++Low
Kalinina, 2015--NANA+--+++++Moderate
Lee, 2006--NANA+--+++++Moderate
Lee, 2012--NANA+--+++++Moderate
Lee, 2015--NANA+--+++++Moderate
Ma, 2014--NANA+--+++++Moderate
Malladi, 2006--NANA+--++++Low
Merceron, 2010--NANA++--+++++High
Park, 2013--NANA++--++++Moderate
Pattapa, 2013++-NANA++--++++High
Russo, 2013--NANA+--++-++Moderate
Salamanna, 2018--NANA+--+-++Low
Sengupta, 2010--NANA+--++++Low
Tsang, 2013--NANA+-+++++Low
Wang, 2012--NANA+--++++Low
Xu, 2007--NANA++--++++Moderate
Xu, 2013--NANA++--++++Moderate
Yang, 2011--NANA+--++++Low
Yao, 2017+-NANA+--++++Low
Zham, 2008--NANA+--++++++Moderate
Zhang, 2017--NANA++--++++Moderate
Zhang, 2018--NANA---+-++Low
++: definitely low; +: probably low; -: probably high (not report); --: definitely high; NA: not applicable. ++ ++ High confidence in the association between exposure to the substance and the outcome. The true effect is highly likely to be reflected in the apparent relationship; + ++ Moderate confidence in the association between exposure to the substance and the outcome. The true effect may be reflected in the apparent relationship; ++ Low confidence in the association between exposure to the substance and the outcome. The true effect may be different from the apparent relationship; + Very low confidence in the association between exposure to the substance and the outcome. The true effect is highly likely to be different from the apparent relationship.

Appendix C

Table A2. Risk of bias and confidence rating ratings for in vivo studies.
Table A2. Risk of bias and confidence rating ratings for in vivo studies.
ReferenceRisk of Bias QuestionsType of StudyConfidence Rating
1234567891011
Wang, 2016+-NANA--++++++++EAHigh
Wang, 2017++-NANA+--+++++EAHigh
Guner, 2013--NANA+-++-+++EAModerate
Tomiyama, 2008NANA+++NANA--++++HCr-SeHigh
Basu, 2013NANA+++NANA++-+++HCr-SeHigh
Basu 2014NANA+++NANA++-+++HCoHigh
Sforza, 2013NANA+++NANA-++++++HCoHigh
Terzi, 2015NANA--++NANA-+++++++HCoHigh
Tng, 2008NANA--++NANA-++++++HCoHigh
O’Brien, 2018--NANANA--+++++HCTModerate
Martínez-Guardado, 2019++++NANANA+++++++++HCTHigh
Ramos-Campos, 2015+-NANANA-++++++++HCTHigh
Rittweger, 2016--NANANA-++++++++HCTHigh
++: definitely low; +: probably low; -: probably high (not report); --: definitely high; NA: not applicable; EA: experimental animal; HCT: human controlled trial; HCo: human cohort; HCr-Se: human cross-sectional. ++ ++ High confidence in the association between exposure to the substance and the outcome: The true effect is highly likely to be reflected in the apparent relationship; + ++ Moderate confidence in the association between exposure to the substance and the outcome: The true effect may be reflected in the apparent relationship; ++ Low confidence in the association between exposure to the substance and the outcome: The true effect may be different from the apparent relationship; + Very low confidence in the association between exposure to the substance and the outcome: The true effect is highly likely to be different from the apparent relationship.

Appendix D

Table A3. Summary risk of bias domain assessment for animal, human and in vitro studies included in the review.
Table A3. Summary risk of bias domain assessment for animal, human and in vitro studies included in the review.
DomainAnimal StudiesHuman StudiesIn Vitro Studies
Selection+++-+-+++-
Performance+--+++-
Attrition/exclusion++-+++-+-
Detection++++++-+++-
Selective Reporting+++++++++
++: definitely low (dark green colour); +: probably low (light green colour); -: probably high (not report; red colour).

Appendix E

Table A4. Confidence rating for a health effect given strengths and weaknesses of a collection of animal and human studies.
Table A4. Confidence rating for a health effect given strengths and weaknesses of a collection of animal and human studies.
Type of StudyLevel of Confidence for Health Effect Bone Remodelling
Experimental AnimalHigh
Human Controlled TrialHigh
Human CohortHigh
Human Cross-SectionalHigh
In Vitro StudiesModerate

References

  1. Dempsey, J.A.; Morgan, B.J. Humans in Hypoxia: A Conspiracy of Maladaptation? Physiology 2015, 30, 304–316. [Google Scholar] [CrossRef] [PubMed]
  2. Dehne, N.; Brune, B. HIF-1 in the inflammatory microenvironment. Exp. Cell Res. 2009, 315, 1791–1797. [Google Scholar] [CrossRef]
  3. Goda, N.; Kanai, M. Hypoxia-inducible factors and their roles in energy metabolism. Int. J. Hematol. 2012, 95, 457–463. [Google Scholar] [CrossRef] [Green Version]
  4. Basovich, S.N. Trends in the use of preconditioning to hypoxia for early prevention of future life diseases. Biosci. Trends 2013, 7, 23–32. [Google Scholar] [CrossRef] [PubMed]
  5. Sadoshima, J. Redox regulation of growth and death in cardiac myocytes. Antioxid. Redox Signal. 2006, 8, 1621–1624. [Google Scholar] [CrossRef] [PubMed]
  6. Almendros, I.; Wang, Y.; Gozal, D. The polymorphic and contradictory aspects of intermittent hypoxia. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 307, L129–L140. [Google Scholar] [CrossRef] [Green Version]
  7. Navarrete-Opazo, A.; Mitchell, G.S. Therapeutic potential of intermittent hypoxia: A matter of dose. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R1181–R1197. [Google Scholar] [CrossRef]
  8. Weiss, M.; Ben-Shlomo, A.B.; Hagag, P.; Rapoport, M. Reference database for bone speed of sound measurement by a novel quantitative multi-site ultrasound device. Osteop. Int. 2000, 11, 688–696. [Google Scholar] [CrossRef] [PubMed]
  9. Beck, B.R.; Norling, T.L. The effect of 8 mos of twice-weekly low- or higher intensity whole body vibration on risk factors for postmenopausal hip fracture. Am. J. Phys. Med. Rehabil. 2010, 89, 997–1009. [Google Scholar] [CrossRef] [PubMed]
  10. Dirckx, N.; Tower, R.J.; Mercken, E.M.; Vangoitsenhoven, R.; Moreau-Triby, C.; Breugelmans, T.; Nefyodova, E.; Cardoen, R.; Mathieu, C.; Van der Schueren, B.; et al. Vhl deletion in osteoblasts boosts cellular glycolysis and improves global glucose metabolism. J. Clin. Investig. 2018, 128, 1087–1105. [Google Scholar] [CrossRef] [PubMed]
  11. Lee, W.C.; Guntur, A.R.; Long, F.; Rosen, C.J. Energy Metabolism of the Osteoblast: Implications for Osteoporosis. Endocr. Rev. 2017, 38, 255–266. [Google Scholar] [CrossRef]
  12. Fan, L.; Li, J.; Yu, Z.; Dang, X.; Wang, K. The hypoxia-inducible factor pathway, prolyl hydroxylase domain protein inhibitors, and their roles in bone repair and regeneration. Biomed. Res. Int. 2014, 2014, 239356. [Google Scholar] [CrossRef]
  13. Serebrovska, T.V.; Serebrovska, Z.O.; Egorov, E. Fitness and therapeutic potential of intermittent hypoxia training: A matter of dose. Fiziol Zh 2016, 62, 78–91. [Google Scholar] [CrossRef]
  14. Xi, L.; Serebrovskaya, T.V. Intermittent Hypoxia and Human Diseases; Springer: London, UK, 2012. [Google Scholar]
  15. Verges, S.; Chacaroun, S.; Godin-Ribuot, D.; Baillieul, S. Hypoxic Conditioning as a New Therapeutic Modality. Front. Pediatrics 2015, 3, 58. [Google Scholar] [CrossRef]
  16. Lavie, L. Obstructive sleep apnoea syndrome—An oxidative stress disorder. Sleep Med. Rev. 2003, 7, 35–51. [Google Scholar] [CrossRef]
  17. Garvey, J.F.; Taylor, C.T.; McNicholas, W.T. Cardiovascular disease in obstructive sleep apnoea syndrome: The role of intermittent hypoxia and inflammation. Eur. Respir J. 2009, 33, 1195–1205. [Google Scholar] [CrossRef]
  18. Urdampilleta, A.; Gonzalez-Muniesa, P.; Portillo, M.P.; Martinez, J.A. Usefulness of combining intermittent hypoxia and physical exercise in the treatment of obesity. J. Physiol. Biochem. 2012, 68, 289–304. [Google Scholar] [CrossRef]
  19. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gotzsche, P.C.; Ioannidis, J.P.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. J. Clin. Epidemiol. 2009, 62, e1–e34. [Google Scholar] [CrossRef]
  20. NTP. Handbook for Conducting a Literature-Based Health Assessment Using OHAT Approach for Systematic Review and Evidence Integration; National Institute of Environmental Health Science: Durham, NC, USA, 2015.
  21. Matta, K.; Ploteau, S.; Coumoul, X.; Koual, M.; Le Bizec, B.; Antignac, J.P.; Cano-Sancho, G. Associations between exposure to organochlorine chemicals and endometriosis in experimental studies: A systematic review protocol. Environ. Int. 2019, 124, 400–407. [Google Scholar] [CrossRef]
  22. Runkle, J.; Flocks, J.; Economos, J.; Dunlop, A.L. A systematic review of Mancozeb as a reproductive and developmental hazard. Environ. Int. 2017, 99, 29–42. [Google Scholar] [CrossRef]
  23. Huang, Y.C.; Zhu, H.M.; Cai, J.Q.; Huang, Y.Z.; Xu, J.; Zhou, Y.; Chen, X.H.; Li, X.Q.; Yang, Z.M.; Deng, L. Hypoxia inhibits the spontaneous calcification of bone marrow-derived mesenchymal stem cells. J. Cell. Biochem. 2012, 113, 1407–1415. [Google Scholar] [CrossRef]
  24. Kalinina, N.; Kharlampieva, D.; Loguinova, M.; Butenko, I.; Pobeguts, O.; Efimenko, A.; Ageeva, L.; Sharonov, G.; Ischenko, D.; Alekseev, D.; et al. Characterization of secretomes provides evidence for adipose-derived mesenchymal stromal cells subtypes. Stem Cell Res. Ther. 2015, 6, 221. [Google Scholar] [CrossRef]
  25. Deschepper, M.; Oudina, K.; David, B.; Myrtil, V.; Collet, C.; Bensidhoum, M.; Logeart-Avramoglou, D.; Petite, H. Survival and function of mesenchymal stem cells (MSCs) depend on glucose to overcome exposure to long-term, severe and continuous hypoxia. J. Cell. Mol. Med. 2011, 15, 1505–1514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gao, Y.S.; Ding, H.; Xie, X.T.; Zhang, C.Q. Osteogenic induction protects rat bone marrow-derived mesenchymal stem cells against hypoxia-induced apoptosis in vitro. J. Surg. Res. 2013, 184, 873–879. [Google Scholar] [CrossRef]
  27. Ding, H.; Chen, S.; Yin, J.H.; Xie, X.T.; Zhu, Z.H.; Gao, Y.S.; Zhang, C.Q. Continuous hypoxia regulates the osteogenic potential of mesenchymal stem cells in a time-dependent manner. Mol. Med. Rep. 2014, 10, 2184–2190. [Google Scholar] [CrossRef]
  28. Lee, S.K.; Gardner, A.E.; Kalinowski, J.F.; Jastrzebski, S.L.; Lorenzo, J.A. RANKL-stimulated osteoclast-like cell formation in vitro is partially dependent on endogenous interleukin-1 production. Bone 2006, 38, 678–685. [Google Scholar] [CrossRef]
  29. Burian, E.; Probst, F.; Palla, B.; Riedel, C.; Saller, M.M.; Cornelsen, M.; Konig, F.; Schieker, M.; Otto, S. Effect of hypoxia on the proliferation of porcine bone marrow-derived mesenchymal stem cells and adipose-derived mesenchymal stem cells in 2- and 3-dimensional culture. J. Cranio-Maxillo-Facial Surg. Off. Publ. Eur. Assoc. Cranio-Maxillo-Facial Surg. 2017, 45, 414–419. [Google Scholar] [CrossRef]
  30. Bouvard, B.; Abed, E.; Yelehe-Okouma, M.; Bianchi, A.; Mainard, D.; Netter, P.; Jouzeau, J.Y.; Lajeunesse, D.; Reboul, P. Hypoxia and vitamin D differently contribute to leptin and dickkopf-related protein 2 production in human osteoarthritic subchondral bone osteoblasts. Arthritis Res. Ther. 2014, 16, 459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Zhang, P.; Ha, N.; Dai, Q.; Zhou, S.; Yu, C.; Jiang, L. Hypoxia suppresses osteogenesis of bone mesenchymal stem cells via the extracellular signalregulated 1/2 and p38mitogen activated protein kinase signaling pathways. Mol. Med. Rep. 2017, 16, 5515–5522. [Google Scholar] [CrossRef] [PubMed]
  32. Salamanna, F.; Cepollaro, S.; Contartese, D.; Giavaresi, G.; Brodano, G.B.; Griffoni, C.; Gasbarrini, A.; Fini, M. Biological Rationale for the Use of Vertebral Whole Bone Marrow in Spinal Surgery. Spine 2018, 43, 1401–1410. [Google Scholar] [CrossRef] [PubMed]
  33. Ciapetti, G.; Granchi, D.; Fotia, C.; Savarino, L.; Dallari, D.; Del Piccolo, N.; Donati, D.M.; Baldini, N. Effects of hypoxia on osteogenic differentiation of mesenchymal stromal cells used as a cell therapy for avascular necrosis of the femoral head. Cytotherapy 2016, 18, 1087–1099. [Google Scholar] [CrossRef]
  34. Tsang, W.P.; Shu, Y.; Kwok, P.L.; Zhang, F.; Lee, K.K.; Tang, M.K.; Li, G.; Chan, K.M.; Chan, W.Y.; Wan, C. CD146+ human umbilical cord perivascular cells maintain stemness under hypoxia and as a cell source for skeletal regeneration. PLoS ONE 2013, 8, e76153. [Google Scholar] [CrossRef]
  35. Gu, Q.; Gu, Y.; Shi, Q.; Yang, H. Hypoxia Promotes Osteogenesis of Human Placental-Derived Mesenchymal Stem Cells. Tohoku J. Exp. Med. 2016, 239, 287–296. [Google Scholar] [CrossRef] [Green Version]
  36. Sengupta, S.; Park, S.H.; Patel, A.; Carn, J.; Lee, K.; Kaplan, D.L. Hypoxia and amino acid supplementation synergistically promote the osteogenesis of human mesenchymal stem cells on silk protein scaffolds. Tissue Eng. Part A 2010, 16, 3623–3634. [Google Scholar] [CrossRef]
  37. Jin, Y.; Kato, T.; Furu, M.; Nasu, A.; Kajita, Y.; Mitsui, H.; Ueda, M.; Aoyama, T.; Nakayama, T.; Nakamura, T.; et al. Mesenchymal stem cells cultured under hypoxia escape from senescence via down-regulation of p16 and extracellular signal regulated kinase. Biochem. Biophys. Res. Commun. 2010, 391, 1471–1476. [Google Scholar] [CrossRef]
  38. Binder, B.Y.; Sagun, J.E.; Leach, J.K. Reduced serum and hypoxic culture conditions enhance the osteogenic potential of human mesenchymal stem cells. Stem Cell Rev. 2015, 11, 387–393. [Google Scholar] [CrossRef]
  39. Xu, Y.; Malladi, P.; Chiou, M.; Bekerman, E.; Giaccia, A.J.; Longaker, M.T. In vitro expansion of adipose-derived adult stromal cells in hypoxia enhances early chondrogenesis. Tissue Eng. 2007, 13, 2981–2993. [Google Scholar] [CrossRef]
  40. Lee, W.Y.; Lui, P.P.; Rui, Y.F. Hypoxia-mediated efficient expansion of human tendon-derived stem cells in vitro. Tissue Eng. Part A 2012, 18, 484–498. [Google Scholar] [CrossRef]
  41. Holzwarth, C.; Vaegler, M.; Gieseke, F.; Pfister, S.M.; Handgretinger, R.; Kerst, G.; Muller, I. Low physiologic oxygen tensions reduce proliferation and differentiation of human multipotent mesenchymal stromal cells. BMC Cell Biol. 2010, 11, 11. [Google Scholar] [CrossRef]
  42. Russo, V.; Yu, C.; Belliveau, P.; Hamilton, A.; Flynn, L.E. Comparison of human adipose-derived stem cells isolated from subcutaneous, omental, and intrathoracic adipose tissue depots for regenerative applications. Stem Cells Transl. Med. 2014, 3, 206–217. [Google Scholar] [CrossRef]
  43. Iacono, E.; Pascucci, L.; Bazzucchi, C.; Cunto, M.; Ricci, F.; Rossi, B.; Merlo, B. Could hypoxia influence basic biological properties and ultrastructural features of adult canine mesenchymal stem/stromal cells? Vet. Res. Commun. 2018, 42, 297–308. [Google Scholar] [CrossRef]
  44. Lee, J.S.; Park, J.C.; Kim, T.W.; Jung, B.J.; Lee, Y.; Shim, E.K.; Park, S.; Choi, E.Y.; Cho, K.S.; Kim, C.S. Human bone marrow stem cells cultured under hypoxic conditions present altered characteristics and enhanced in vivo tissue regeneration. Bone 2015, 78, 34–45. [Google Scholar] [CrossRef]
  45. Hsu, S.H.; Chen, C.T.; Wei, Y.H. Inhibitory effects of hypoxia on metabolic switch and osteogenic differentiation of human mesenchymal stem cells. Stem Cells 2013, 31, 2779–2788. [Google Scholar] [CrossRef]
  46. Park, I.H.; Kim, K.H.; Choi, H.K.; Shim, J.S.; Whang, S.Y.; Hahn, S.J.; Kwon, O.J.; Oh, I.H. Constitutive stabilization of hypoxia-inducible factor alpha selectively promotes the self-renewal of mesenchymal progenitors and maintains mesenchymal stromal cells in an undifferentiated state. Exp. Mol. Med. 2013, 45, e44. [Google Scholar] [CrossRef]
  47. Ma, H.P.; Ma, X.N.; Ge, B.F.; Zhen, P.; Zhou, J.; Gao, Y.H.; Xian, C.J.; Chen, K.M. Icariin attenuates hypoxia-induced oxidative stress and apoptosis in osteoblasts and preserves their osteogenic differentiation potential in vitro. Cell Prolif. 2014, 47, 527–539. [Google Scholar] [CrossRef]
  48. Yao, Y.; Deng, Q.; Sun, C.; Song, W.; Liu, H.; Zhou, Y. A genome-wide analysis of the gene expression profiles and alternative splicing events during the hypoxia-regulated osteogenic differentiation of human cartilage endplate-derived stem cells. Mol. Med. Rep. 2017, 16, 1991–2001. [Google Scholar] [CrossRef] [Green Version]
  49. Xu, N.; Liu, H.; Qu, F.; Fan, J.; Mao, K.; Yin, Y.; Liu, J.; Geng, Z.; Wang, Y. Hypoxia inhibits the differentiation of mesenchymal stem cells into osteoblasts by activation of Notch signaling. Exp. Mol. Pathol. 2013, 94, 33–39. [Google Scholar] [CrossRef]
  50. Yang, D.C.; Yang, M.H.; Tsai, C.C.; Huang, T.F.; Chen, Y.H.; Hung, S.C. Hypoxia inhibits osteogenesis in human mesenchymal stem cells through direct regulation of RUNX2 by TWIST. PLoS ONE 2011, 6, e23965. [Google Scholar] [CrossRef]
  51. Cicione, C.; Muinos-Lopez, E.; Hermida-Gomez, T.; Fuentes-Boquete, I.; Diaz-Prado, S.; Blanco, F.J. Effects of severe hypoxia on bone marrow mesenchymal stem cells differentiation potential. Stem Cells Int. 2013, 2013, 232896. [Google Scholar] [CrossRef]
  52. Pattappa, G.; Thorpe, S.D.; Jegard, N.C.; Heywood, H.K.; de Bruijn, J.D.; Lee, D.A. Continuous and uninterrupted oxygen tension influences the colony formation and oxidative metabolism of human mesenchymal stem cells. Tissue Eng. Part Cmethods 2013, 19, 68–79. [Google Scholar] [CrossRef]
  53. Zahm, A.M.; Bucaro, M.A.; Srinivas, V.; Shapiro, I.M.; Adams, C.S. Oxygen tension regulates preosteocyte maturation and mineralization. Bone 2008, 43, 25–31. [Google Scholar] [CrossRef] [Green Version]
  54. Malladi, P.; Xu, Y.; Chiou, M.; Giaccia, A.J.; Longaker, M.T. Effect of reduced oxygen tension on chondrogenesis and osteogenesis in adipose-derived mesenchymal cells. Am. J. Physiol. Cell Physiol. 2006, 290, C1139–C1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Inagaki, Y.; Akahane, M.; Shimizu, T.; Inoue, K.; Egawa, T.; Kira, T.; Ogawa, M.; Kawate, K.; Tanaka, Y. Modifying oxygen tension affects bone marrow stromal cell osteogenesis for regenerative medicine. World J. Stem Cells 2017, 9, 98–106. [Google Scholar] [CrossRef]
  56. Hopper, N.; Wardale, J.; Brooks, R.; Power, J.; Rushton, N.; Henson, F. Peripheral Blood Mononuclear Cells Enhance Cartilage Repair in in vivo Osteochondral Defect Model. PLoS ONE 2015, 10, e0133937. [Google Scholar] [CrossRef] [PubMed]
  57. Merceron, C.; Vinatier, C.; Portron, S.; Masson, M.; Amiaud, J.; Guigand, L.; Cherel, Y.; Weiss, P.; Guicheux, J. Differential effects of hypoxia on osteochondrogenic potential of human adipose-derived stem cells. Am. J. Physiol. Cell Physiol. 2010, 298, C355–C364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. D’Ippolito, G.; Diabira, S.; Howard, G.A.; Roos, B.A.; Schiller, P.C. Low oxygen tension inhibits osteogenic differentiation and enhances stemness of human MIAMI cells. Bone 2006, 39, 513–522. [Google Scholar] [CrossRef] [PubMed]
  59. Wang, W.; Yun, Z.; Peng, H.; Yan, S.; Zhang, H.; Qiu, X.; Wen, Y.; Long, H.; Ma, B. The hypobaric hypoxia environment impairs bone strength and quality in rats. Int J. Clin. Exp. Med. 2017, 10, 9397–9406. [Google Scholar]
  60. O’Brien, K.A.; Pollock, R.D.; Stroud, M.; Lambert, R.J.; Kumar, A.; Atkinson, R.A.; Green, D.A.; Anton-Solanas, A.; Edwards, L.M.; Harridge, S.D.R. Human physiological and metabolic responses to an attempted winter crossing of Antarctica: The effects of prolonged hypobaric hypoxia. Physiol. Rep. 2018, 6, e13613. [Google Scholar] [CrossRef]
  61. Basu, M.; Malhotra, A.S.; Pal, K.; Kumar, R.; Bajaj, R.; Verma, S.K.; Ghosh, D.; Sharma, Y.K.; Sawhney, R.C. Alterations in Different Indices of Skeletal Health after Prolonged Residency at High Altitude. High. Alt. Med. Biol. 2014, 15, 170–175. [Google Scholar] [CrossRef]
  62. Basu, M.; Malhotra, A.S.; Pal, K.; Chatterjee, T.; Ghosh, D.; Haldar, K.; Verma, S.K.; Kumar, S.; Sharma, Y.K.; Sawhney, R.C. Determination of bone mass using multisite quantitative ultrasound and biochemical markers of bone turnover during residency at extreme altitude: A longitudinal study. High. Alt. Med. Biol 2013, 14, 150–154. [Google Scholar] [CrossRef]
  63. Wang, G.; Wang, J.; Sun, D.; Xin, J.; Wang, L.; Huang, D.; Wu, W.; Xian, C.J. Short-Term Hypoxia Accelerates Bone Loss in Ovariectomized Rats by Suppressing Osteoblastogenesis but Enhancing Osteoclastogenesis. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2016, 22, 2962–2971. [Google Scholar] [CrossRef] [Green Version]
  64. Guner, I.; Uzun, D.D.; Yaman, M.O.; Genc, H.; Gelisgen, R.; Korkmaz, G.G.; Hallac, M.; Yelmen, N.; Sahin, G.; Karter, Y.; et al. The effect of chronic long-term intermittent hypobaric hypoxia on bone mineral density in rats: Role of nitric oxide. Biol. Trace Elem. Res. 2013, 154, 262–267. [Google Scholar] [CrossRef]
  65. Martinez-Guardado, I.; Ramos-Campo, D.J.; Olcina, G.J.; Rubio-Arias, J.A.; Chung, L.H.; Marin-Cascales, E.; Alcaraz, P.E.; Timon, R. Effects of high-intensity resistance circuit-based training in hypoxia on body composition and strength performance. Eur. J. Sport Sci. 2019, 1–11. [Google Scholar] [CrossRef]
  66. Ramos-Campo, D.J.; Rubio-Arias, J.A.; Jimenez-Diaz, J.F. Effects in body composition and bone mineral density of simulate altitude program in triathletes. Nutr. Hosp. 2015, 32, 1252–1260. [Google Scholar]
  67. Rittweger, J.; Debevec, T.; Frings-Meuthen, P.; Lau, P.; Mittag, U.; Ganse, B.; Ferstl, P.G.; Simpson, E.J.; Macdonald, I.A.; Eiken, O.; et al. On the combined effects of normobaric hypoxia and bed rest upon bone and mineral metabolism: Results from the PlanHab study. Bone 2016, 91, 130–138. [Google Scholar] [CrossRef] [Green Version]
  68. Tng, H.Y.; Thu, W.P.P.; Logan, S.; Aris, I.M.; Cauley, J.; Yong, E.L. Sleep apnea and femoral neck BMD among Singaporean mid-life women. Arch. Osteoporos. 2018, 13, 19. [Google Scholar] [CrossRef]
  69. Terzi, R.; Yilmaz, Z. Bone mineral density and changes in bone metabolism in patients with obstructive sleep apnea syndrome. J. Bone Miner. Metab. 2016, 34, 475–481. [Google Scholar] [CrossRef]
  70. Sforza, E.; Thomas, T.; Barthelemy, J.C.; Collet, P.; Roche, F. Obstructive sleep apnea is associated with preserved bone mineral density in healthy elderly subjects. Sleep 2013, 36, 1509–1515. [Google Scholar]
  71. Tomiyama, H.; Okazaki, R.; Inoue, D.; Ochiai, H.; Shiina, K.; Takata, Y.; Hashimoto, H.; Yamashina, A. Link between obstructive sleep apnea and increased bone resorption in men. Osteoporos. Int. A J. Establ. Result Coop. Between Eur. Found. Osteoporos. Natl. Osteoporos. Found. USA 2008, 19, 1185–1192. [Google Scholar] [CrossRef]
  72. Wang, Y.; Li, J.; Wang, Y.; Lei, L.; Jiang, C.; An, S.; Zhan, Y.; Cheng, Q.; Zhao, Z.; Wang, J.; et al. Effects of hypoxia on osteogenic differentiation of rat bone marrow mesenchymal stem cells. Mol. Cell. Biochem. 2012, 362, 25–33. [Google Scholar] [CrossRef]
  73. Mateika, J.H.; El-Chami, M.; Shaheen, D.; Ivers, B. Intermittent hypoxia: A low-risk research tool with therapeutic value in humans. J. Appl. Physiol. 2015, 118, 520–532. [Google Scholar] [CrossRef]
  74. Barcelo, A.; Pierola, J.; de la Pena, M.; Esquinas, C.; Fuster, A.; Sanchez-de-la-Torre, M.; Carrera, M.; Alonso-Fernandez, A.; Ladaria, A.; Bosch, M.; et al. Free fatty acids and the metabolic syndrome in patients with obstructive sleep apnoea. Eur. Respir. J. 2011, 37, 1418–1423. [Google Scholar] [CrossRef]
  75. Destors, M.; Tamisier, R.; Baguet, J.P.; Levy, P.; Pepin, J.L. Cardiovascular morbidity associated with obstructive sleep apnea syndrome. Rev. Mal. Respir. 2014, 31, 375–385. [Google Scholar] [CrossRef]
  76. Huang, J.; Deng, F.; Wang, L.; Xiang, X.R.; Zhou, W.W.; Hu, N.; Xu, L. Hypoxia induces osteogenesis-related activities and expression of core binding factor alpha1 in mesenchymal stem cells. Tohoku J. Exp. Med. 2011, 224, 7–12. [Google Scholar] [CrossRef]
  77. Zhang, J.; Feng, Z.; Wei, J.; Yu, Y.; Luo, J.; Zhou, J.; Li, Y.; Zheng, X.; Tang, W.; Liu, L.; et al. Repair of Critical-Sized Mandible Defects in Aged Rat Using Hypoxia Preconditioned BMSCs with Up-regulation of Hif-1alpha. Int. J. Biol. Sci. 2018, 14, 449–460. [Google Scholar] [CrossRef]
  78. Lee, J.H.; Kemp, D.M. Human adipose-derived stem cells display myogenic potential and perturbed function in hypoxic conditions. Biochem. Biophys. Res. Commun. 2006, 341, 882–888. [Google Scholar] [CrossRef]
  79. Hung, S.P.; Ho, J.H.; Shih, Y.R.; Lo, T.; Lee, O.K. Hypoxia promotes proliferation and osteogenic differentiation potentials of human mesenchymal stem cells. J. Orthop. Res. Off. Publ. Orthop. Res. Soc. 2012, 30, 260–266. [Google Scholar] [CrossRef]
  80. Bailey, S.; Karsenty, G.; Gundberg, C.; Vashishth, D. Osteocalcin and osteopontin influence bone morphology and mechanical properties. Ann. N. Y. Acad. Sci. 2017, 1409, 79–84. [Google Scholar] [CrossRef]
  81. Ishijima, M.; Rittling, S.R.; Yamashita, T.; Tsuji, K.; Kurosawa, H.; Nifuji, A.; Denhardt, D.T.; Noda, M. Enhancement of osteoclastic bone resorption and suppression of osteoblastic bone formation in response to reduced mechanical stress do not occur in the absence of osteopontin. J. Exp. Med. 2001, 193, 399–404. [Google Scholar] [CrossRef]
  82. Dos Santos, F.; Andrade, P.Z.; Boura, J.S.; Abecasis, M.M.; da Silva, C.L.; Cabral, J.M. Ex vivo expansion of human mesenchymal stem cells: A more effective cell proliferation kinetics and metabolism under hypoxia. J. Cell Physiol. 2010, 223, 27–35. [Google Scholar] [CrossRef]
  83. Zhang, Q.B.; Zhang, Z.Q.; Fang, S.L.; Liu, Y.R.; Jiang, G.; Li, K.F. Effects of hypoxia on proliferation and osteogenic differentiation of periodontal ligament stem cells: An in vitro and in vivo study. Genet. Mol. Res. 2014, 13, 10204–10214. [Google Scholar] [CrossRef]
  84. Khan, W.S.; Adesida, A.B.; Tew, S.R.; Lowe, E.T.; Hardingham, T.E. Bone marrow-derived mesenchymal stem cells express the pericyte marker 3G5 in culture and show enhanced chondrogenesis in hypoxic conditions. J. Orthop. Res. Off. Publ. Orthop. Res. Soc. 2010, 28, 834–840. [Google Scholar] [CrossRef]
  85. Martin-Rendon, E.; Hale, S.J.; Ryan, D.; Baban, D.; Forde, S.P.; Roubelakis, M.; Sweeney, D.; Moukayed, M.; Harris, A.L.; Davies, K.; et al. Transcriptional profiling of human cord blood CD133+ and cultured bone marrow mesenchymal stem cells in response to hypoxia. Stem Cells 2007, 25, 1003–1012. [Google Scholar] [CrossRef]
  86. Gordillo, G.M.; Sen, C.K. Revisiting the essential role of oxygen in wound healing. Am. J. Surg. 2003, 186, 259–263. [Google Scholar] [CrossRef]
  87. Utting, J.C.; Flanagan, A.M.; Brandao-Burch, A.; Orriss, I.R.; Arnett, T.R. Hypoxia stimulates osteoclast formation from human peripheral blood. Cell Biochem. Funct. 2010, 28, 374–380. [Google Scholar] [CrossRef]
  88. Arnett, T.R.; Gibbons, D.C.; Utting, J.C.; Orriss, I.R.; Hoebertz, A.; Rosendaal, M.; Meghji, S. Hypoxia is a major stimulator of osteoclast formation and bone resorption. J. Cell Physiol. 2003, 196, 2–8. [Google Scholar] [CrossRef] [Green Version]
  89. Qin, L.; Raggatt, L.J.; Partridge, N.C. Parathyroid hormone: A double-edged sword for bone metabolism. Trends Endocrinol. Metab. Tem. 2004, 15, 60–65. [Google Scholar] [CrossRef]
  90. Bingham, P.J.; Brazell, I.A.; Owen, M. The effect of parathyroid extract on cellular activity and plasma calcium levels in vivo. J. Endocrinol. 1969, 45, 387–400. [Google Scholar] [CrossRef]
  91. Bozzini, C.E.; Lezon, C.E.; Norese, M.F.; Conti, M.I.; Martinez, M.P.; Olivera, M.I.; Alippi, R.M. Evidence from catch-up growth and hoarding behavior of rats that exposure to hypobaric air lowers the body-mass set point. Growth Dev. Aging 2005, 69, 81–88. [Google Scholar]
  92. Vuori, I.M. Dose-response of physical activity and low back pain, osteoarthritis, and osteoporosis. Med. Sci. Sports Exerc. 2001, 33 (Suppl. 6), S551–S586. [Google Scholar] [CrossRef] [Green Version]
  93. Kelly, L.P.; Basset, F.A. Acute Normobaric Hypoxia Increases Post-exercise Lipid Oxidation in Healthy Males. Front. Physiol. 2017, 8, 293. [Google Scholar] [CrossRef] [Green Version]
  94. Aghajanian, P.; Hall, S.; Wongworawat, M.D.; Mohan, S. The Roles and Mechanisms of Actions of Vitamin C in Bone: New Developments. J. Bone Miner. Res. Off. J. Am. Soc. Bone Mineral. Res. 2015, 30, 1945–1955. [Google Scholar] [CrossRef] [Green Version]
  95. Bischoff-Ferrari, H.A.; Dietrich, T.; Orav, E.J.; Dawson-Hughes, B. Positive association between 25-hydroxy vitamin D levels and bone mineral density: A population-based study of younger and older adults. Am. J. Med. 2004, 116, 634–639. [Google Scholar] [CrossRef]
  96. Kent, B.D.; Mitchell, P.D.; McNicholas, W.T. Hypoxemia in patients with COPD: Cause, effects, and disease progression. Int. J. Chron Obs. Pulmon Dis. 2011, 6, 199–208. [Google Scholar]
  97. Knapp, K.M.; Blake, G.M.; Spector, T.D.; Fogelman, I. Multisite quantitative ultrasound: Precision, age- and menopause-related changes, fracture discrimination, and T-score equivalence with dual-energy X-ray absorptiometry. Osteoporos. Int. A J. Establ. Result Coop. Between Eur. Found. Osteoporos. Natl. Osteoporos. Found. USA 2001, 12, 456–464. [Google Scholar] [CrossRef]
  98. Litovka, I.H. Alimentary and oxygen deprivation as the modulator of the bone tissue physiological remodelling rate in young rats. Fiziol. Zh. 2008, 54, 85–93. [Google Scholar]
  99. Swanson, C.M.; Shea, S.A.; Stone, K.L.; Cauley, J.A.; Rosen, C.J.; Redline, S.; Karsenty, G.; Orwoll, E.S. Obstructive sleep apnea and metabolic bone disease: Insights into the relationship between bone and sleep. J. Bone Miner. Res. Off. J. Am. Soc. Bone Mineral. Res. 2015, 30, 199–211. [Google Scholar] [CrossRef]
  100. Orriss, I.R.; Knight, G.E.; Utting, J.C.; Taylor, S.E.; Burnstock, G.; Arnett, T.R. Hypoxia stimulates vesicular ATP release from rat osteoblasts. J. Cell Physiol. 2009, 220, 155–162. [Google Scholar] [CrossRef]
Figure 1. Flowchart of article searches and selection strategies.
Figure 1. Flowchart of article searches and selection strategies.
Ijerph 16 01799 g001
Table 1. Experimental details of in vitro studies included in this review.
Table 1. Experimental details of in vitro studies included in this review.
Hypoxia Effects on OutcomesHypoxia Level (% PiO2)Duration, FrequencyReferencesConfidence Rating
Sustained Exposure
RUNX2-g
ALP-g
ALP
ALP-activity
Col1A1-g
Col1A1
Osteocalcin-g
Osteocalcin
Calcium deposit
0.1%1 daysHuang, 2012 [23] Moderate
1%2 daysKalinina, 2015 [24] Moderate
12 daysDeschepper, 2011 [25] Moderate
21 daysGao, 2013 [26]
Ding, 2014 [27]
Moderate
High
2%NRLee, 2006 [28]
Burin, 2017 [29]
Moderate
Low
1 dayBouvard, 2014 [30] Moderate
2 daysZhang, 2018 [31] Low
3 daysSalamanna, 2018 [32] Low
12 daysCiapetti, 2016 [33] High
14 daysTsang, 2013 [34] Low
21 daysTsang, 2013 [34] Low
5%3 daysGu, 2016 [35] Moderate
14 daysDing, 2014 [27] High
49 daysSengupta, 2010 [36] Low
RUNX2-g
ALP-g
ALP
ALP-activity
Col1A1-g
Col1A1
Osteocalcin
Osteopontin-g
Osteopontin
Calcium deposit
1%14 daysJin, 2010 [37] Low
21 daysBinder, 2015 [38]
Ding, 2014 [27]
Moderate
High
2%5 daysXu, 2007 [39] Moderate
8 daysXu, 2007 [39] Moderate
12 daysCiapetti, 2016 [33] High
14 daysTsang, 2013 [34]
Zhang, 2017 [31]
Low
Moderate
21 daysTsang, 2013 [34]
Lee, 2012 [40]
Low
Moderate
3%14 daysHolzwarth, 2010 [41] Low
5%NRRusso, 2014 [42] Moderate
3 daysGu, 2016 [35] Moderate
21 daysBinder, 2015 [38] Moderate
49 daysSengupta, 2010 [36] Low
7%NRIacono, 2018 [43] Low
RUNX2-g
RUNX22
ALP-g
ALP
ALP-activity
Col1A1-g
Col1A1
Osteocalcin-g
Osteocalcin
Osteopontin-g
Osteopontin
Calcium deposit
1%NRLee, 2015 [44]
Hsu, 2013 [45]
Park, 2013 [46]
Moderate
Low
Moderate
2 daysMa, 2014 [47] Moderate
21 daysDing, 2014 [27]
Yao, 2017 [48]
Xu, 2013 [49]
Yang, 2011 [50]
Cicione, 2013 [51]
High
Low
Moderate
Low
Moderate
2%NRBurian, 2017 [29] Low
3 daysSalamanna, 2018 [32] Low
5 daysXu, 2007 [39]
Huang, 2012 [23]
Moderate
High
6 daysPattappa, 2013 [52] Moderate
7 daysZham, 2008 [53] Moderate
8 daysXu, 2007 [39] High
14 daysZhang, 2017 [31] Moderate
21 daysHuang, 2012 [23]
Tsang, 2013 [34]
Malladi, 2006 [54]
Lee, 2012 [40]
Moderate
Low
Low
Moderate
3%14 daysHolzwarth, 2010 [41] Low
5%NRRusso, 2014 [42] Moderate
6 daysPattappa, 2013 [52] High
14 daysInagaki, 2017 [55] High
21 daysHopper, 2015 [56] Low
28 daysMerceron, 2010 [57] High
Cyclic Exposure
ALP activity
Calcium deposit
ALP
RUNX2
Osteocalcin
1%
3%
5%
10%
15 days
2 × 3 min/day
Dìppolito, 2006 [58]Moderate
NR: not reported; -g: genetic expression; RUNX2: runt-related transcription factor 2; ALP: alkaline phosphatase; Col1A1: collagen type 1 alpha1. High confidence in the association between exposure to the substance and the outcome: The true effect is highly likely to be reflected in the apparent relationship. Moderate confidence in the association between exposure to the substance and the outcome: The true effect may be reflected in the apparent relationship. Low confidence in the association between exposure to the substance and the outcome: The true effect may be different from the apparent relationship. Very low confidence in the association between exposure to the substance and the outcome: The true effect is highly likely to be different from the apparent relationship.
Table 2. Experimental details of in vivo studies included in this review
Table 2. Experimental details of in vivo studies included in this review
Hypoxia Effects on OutcomesSampleInterventionReferencesConfidence Rating
TypeAgeSizeConditions (n)Hypoxia Level (Meters; % PiO2)Duration, FrequencyExposure Type
Sustained Exposure
BV/TV
Tb.N
BMD-total
Sprague-Dawley rats12 week-oldHypoxia (n = 4)
Normoxia (n = 4)
6000 m3 weeksNormobaric Wang, 2017 [59] High
BMD-spineHealthy adults24–58 years5NR2500 m24 weeksHypobaricO´Brien, 2019
[60]
Moderate
SOS-R
SOS-T
SOS-M
SOS-P
T-score-R
T-score-P
ALP
BAP
Calcium deposit
25-Vit D
i-PTH
CICP
NTX
DPD/Cr
Healthy adults21–47 years2600Normoxia (n = 1300)
Hypoxia (n = 1300)
3450 m16 weeksHypobaricBasu, 2014
[61]
High
SOS-R
SOS-T
SOS-M
SOS-P
Z-score-R
Z-score-T
Z-score-M
Z-score-P
Calcium
Phosphorous
ALP
BAP
25-Vit D
Calcitonin
i-PTH
DPD/Cr
Healthy adults21–47 years221Hypoxia (n = 221)3000–3754 m
(24 weeks)
+ 5400–6700 m
(16 weeks)
40 weeksHypobaricBasu, 2013
[62]
High
Cyclic Exposure
BV/TV
Tb.N
BMD-total
BMC-total
BV/TV
Tb.N
BMD-total
BMC-total
Sprague-Dawley rats12 week-old37Hypoxia (n = 7)
Normoxia (n = 6)
Ovariectomized
Hypoxia (n = 12)
Ovariectomized
Normoxia (n = 12)
3000–5000 m2 weeks,
4 h/day
Normobaric Wang, 2016
[63]
High
BMD-spineWistar albino rats6 months-old20Hypoxia (n = 10)
Normoxia (n = 10)
4500 m5 weeks
5 days/week 5 h/day
Hypobaric Guner, 2013
[64]
Moderate
BMD-totalHealthy adults24.6 ± 2.8 years28Hypoxia (n = 15)
Normoxia (n = 13)
15% PiO28 weeks
2days/week
NormobaricMartínez-Guardado, 2019 [65] High
BMD-totalTrained triathletes27 years18Hypoxia Training (n = 9)
Control (n = 9)
15% PiO27 weeks, 2days/week
60 min/day
NormobaricRamos-Campos, 2015
[66]
High
BMC-total
BMC-total
Healthy young26.4 years14Hypoxia Bed Rest (n = 14)
Hypoxia Ambulatory (n = 14)
Normoxia Bed Rest (n = 14)
4000 m21 daysNormobaricRittweger, 2016 [67] High
Intermittent Exposure
BMD-spineMenopausal Women with OSAS56.3 ± 6.2 years1201NRNRNRNRTng, 2018
[68]
High
CTXAdults with OSAS51 years50OSA (n = 30)
Control (n = 20)
NRNRNRTerzi, 2016
[69]
High
BMD-totalAdults with OSAS68.6 ± 0.8 years833OSA (n = 459)
Control (n = 373)
NRNRNRSforza, 2013
[70]
High
CTX
RANKL
OPG
CTX
RANKL
OPG
CTX
RANKL
OPG
Adults with OSAS51.0 ± 13 years65Mild OSAS (n = 10)
Moderate OSAS (n = 12)
Severe OSAS (n = 28)
Control (n = 15)
NRNRNRTomiyama, 2008
[71]
High
NR: not report; OSAS: obstructive sleep apnea syndrome; SOS-R: speed of sound at one-third of distal radius; SOS-P: speed of sound at the proximal third phalanx; SOS-M: speed of sound at fifth metatarsal; SOS-T: speed of sound at the mid-shaft tibia; BAP: bone specific alkaline phosphatase; 25-Vit D: 25-Hydroxy vitamin D3; i-PTH: intact parathyroid hormone; CICP: C-terminal propeptide of type I collagen; NTX: N-telopeptide of type I collagen; DPD/Cr: urinary DPD, creatinine ratio; BV/TV: bone volume; Tb.N: trabecular number; BMD- total: bone mineral density total; BMD- spine: bone mineral density dorsal spine; BMC-total: bone mineral content total; CTX: carboxy-terminal collagen cross-links; RANKL: Receptor activator for Nuclear Factor κ B Ligand; OPG: osteoprotegerin. High confidence in the association between exposure to the substance and the outcome: The true effect is highly likely to be reflected in the apparent relationship. Moderate confidence in the association between exposure to the substance and the outcome: The true effect may be reflected in the apparent relationship. Low confidence in the association between exposure to the substance and the outcome: The true effect may be different from the apparent relationship. Very low confidence in the association between exposure to the substance and the outcome: The true effect is highly likely to be different from the apparent relationship.

Share and Cite

MDPI and ACS Style

Camacho-Cardenosa, M.; Camacho-Cardenosa, A.; Timón, R.; Olcina, G.; Tomas-Carus, P.; Brazo-Sayavera, J. Can Hypoxic Conditioning Improve Bone Metabolism? A Systematic Review. Int. J. Environ. Res. Public Health 2019, 16, 1799. https://doi.org/10.3390/ijerph16101799

AMA Style

Camacho-Cardenosa M, Camacho-Cardenosa A, Timón R, Olcina G, Tomas-Carus P, Brazo-Sayavera J. Can Hypoxic Conditioning Improve Bone Metabolism? A Systematic Review. International Journal of Environmental Research and Public Health. 2019; 16(10):1799. https://doi.org/10.3390/ijerph16101799

Chicago/Turabian Style

Camacho-Cardenosa, Marta, Alba Camacho-Cardenosa, Rafael Timón, Guillermo Olcina, Pablo Tomas-Carus, and Javier Brazo-Sayavera. 2019. "Can Hypoxic Conditioning Improve Bone Metabolism? A Systematic Review" International Journal of Environmental Research and Public Health 16, no. 10: 1799. https://doi.org/10.3390/ijerph16101799

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

Article Metrics

Back to TopTop