Skip to main content
Erschienen in: Journal of Translational Medicine 1/2014

Open Access 01.12.2014 | Methodology

Combined intermittent hypobaric hypoxia and muscle electro-stimulation: a method to increase circulating progenitor cell concentration?

verfasst von: Luisa Corral, Casimiro Javierre, Juan Blasi, Ginés Viscor, Antoni Ricart, Josep Lluis Ventura

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2014

Abstract

Background

Our goal was to test whether short-term intermittent hypobaric hypoxia (IHH) at a level well tolerated by healthy humans could, in combination with muscle electro-stimulation (ME), mobilize circulating progenitor cells (CPC) and increase their concentration in peripheral circulation.

Methods

Nine healthy male subjects were subjected, as the active group (HME), to a protocol involving IHH plus ME. IHH exposure consisted of four, three-hour sessions at a barometric pressure of 540 hPa (equivalent to an altitude of 5000 m). These sessions took place on four consecutive days. ME was applied in two separate 20-minute periods during each IHH session. Blood samples were obtained from an antecubital vein on three consecutive days immediately before the experiment, and then 24 h, 48 h, 4 days, 7 days and 14 days after the last day of hypoxic exposure. Four months later a control study was carried out involving seven of the original subjects (CG), who underwent the same protocol of blood samples but without receiving any special stimulus.

Results

In comparison with the CG the HME group showed only a non-significant increase in the number of CPC CD34+ cells on the fourth day after the combined IHH and ME treatment.

Conclusion

CPC levels oscillated across the study period and provide no firm evidence to support an increased CPC count after IHH plus ME, although it is not possible to know if this slight increase observed is physiologically relevant. Further studies are required to understand CPC dynamics and the physiology and physiopathology of the hypoxic stimulus.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1479-5876-12-174) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no conflict of interest to disclose.

Authors’ contributions

All six authors have contributed to the study in accordance with the international consensus on authorship, and they have approved the final draft submitted. LC contributed to the design and implementation of the study, to data collection and analysis, and to manuscript writing. CJ contributed to the conception and design of the study, to its implementation, to data collection and analysis, and to manuscript writing. JB contributed to implementation of the study, performed the measurements of circulating progenitor cells, and contributed to data analysis and manuscript writing. GV contributed to the conception and design of the study, to its implementation, to data collection and analysis, and to manuscript writing. AR contributed to the design and implementation of the study and to manuscript writing. JV made a significant contribution to the conception and design of the study, as well as to data analysis and manuscript writing. All authors read and approved the final manuscript.

Background

Human circulating progenitor cells (CPC) have generally been defined as circulating cells that express a variety of cell surface markers similar to those expressed by vascular endothelial cells, that adhere to endothelium at sites of hypoxia/ischemia, and which participate in new vessel formation, contributing to the maintenance of endothelial function and organ perfusion through mechanisms that range from endothelial repair to neovasculogenesis[13]. Several studies have found that elevated concentrations of CPC correlate with better clinical outcomes[4]. An increase in CPC has been observed after various events including myocardial infarction[5], dilated myocardiopathy[6], cardiac surgery with cardiopulmonary bypass[7], 12 weeks of physical exercise[8, 9], menstruation[10], cessation of smoking[11] and traumatic brain injury (TBI)[12], as well as in animal or human cells subjected to deep hypoxia conditions in vitro[1316].
Hypoxia enhances proliferation and tissue formation derived from mesenchymal stem cells in human bone marrow[14]. Functional benefits of repetitive hypoxia have been explored not only for their therapeutic value in patients but also as regards performance improvement in athletes[17]. Above 2500 m of altitude, hypobaric hypoxia elicits different adaptive responses and may also cause certain diseases such as acute mountain sickness, an outcome that depends mainly on the ascent speed and the time spent at altitude[18]. However, when producing intermittent hypobaric hypoxia (IHH) in a hypobaric chamber it is possible to control the temporal parameters equivalent to ascent time, altitude time and descent time, thereby greatly reducing the risk of complications by limiting and tailoring the exposure time to each target and each patient’s tolerance.
Research also suggests that exercise may influence the mobilization of CPC, and probably tissue homing as well, both in healthy people[1923] and in cardiovascular or renal patients[2426]. Although the different forms of standard physical exercise may be difficult to implement under hypoxic conditions, muscle electro-stimulation (ME) is easier to apply and has been shown to be efficient in mimicking training effects and increasing CPC[2729].
A recent study by our group suggested that a short protocol of exposure to IHH in a hypobaric chamber with simultaneous ME is able to increase the concentration of CPC in peripheral blood in humans[29]. This promising finding raises the hope that an increase in CPC could help in the recovery from many diseases. Previous studies have used a variety of protocols to assess CPC mobilization[1726], but their results are not conclusive. A better understanding of the issue in healthy humans may help guide the design of clinical applications and thus aid recovery from a variety of diseases.
The present research sought to reproduce our previous study with a larger sample size, more post-stimulus blood samples and the inclusion of a control group, the aim being to determine whether short-term IHH at a level well tolerated by healthy humans could, in combination with ME, increase CPC.

Methods

Nine healthy males were subjected to a protocol involving IHH plus ME, hypoxic muscle electro-stimulation as the active group (HME). Their median age was 29 years [interquartile range (IQR) 9]. Six of them performed regular athletic exercise (three fewer than 3 times/week and three more than 3 times/week) and one was a smoker. Median height was 176 cm (IQR 11.5), mean weight was 80 kg (IQR 14) and median BMI was 25 (IQR 3).
IHH exposure consisted of four, three-hour sessions at a barometric pressure of 540 hPa (equivalent to an altitude of 5000 m). These sessions were conducted on four consecutive days in the hypobaric chamber of the University of Barcelona at temperature between 24.1 and 26.7°C and relative humidity between 32 and 39%. During the IHH sessions ME was applied using the Compex Vitality® vascular and capillarization program, with electrodes being fixed to the quadriceps and abdominal muscles[30]. ME was applied at the maximal tolerated intensity (regulated individually by each experimental subject) in two separate 20-minute periods during each IHH session.
Blood samples were obtained from an antecubital vein on three consecutive days immediately before the experiment, and then 24 h, 48 h, 4 days, 7 days and 14 days after the last day of hypoxic exposure.
A control study was conducted four months later with seven of these same participants (CG). On this occasion they did not receive any special stimulus, neither IHH nor ME was used, but blood samples were obtained according to the same schedule and on the same days of the week.

Blood sampling, CD34 staining and flow cytometry assay

All blood samples were obtained in the morning and following the same extraction methodology, as detailed below. Peripheral blood samples were collected by puncture of an antecubital vein and deposited into tubes treated with 0.34 M di-potassium ethylenediaminetetraacetic acid anticoagulant. All samples were stored at room temperature and processed within six hours of arrival at the laboratory. Samples were incubated for cytometric absolute count with anti-human fluorescein isothiocyanate (FITC)-conjugated CD45 monoclonal antibody (BD Bioscience) and anti-human phycoerythrin (PE)-conjugated anti-CD34 (BD Bioscience) for 15 min at room temperature. Red blood cells were lysed with 1 ml of quick lysis solution (CYT-QL-1, Cytognos) for 15 min at room temperature. Samples were incubated under dark conditions and analysed immediately. To ensure accuracy, reverse pipetting was used to dispense the volumes.
A single-platform protocol with Perfect-Count microspheres (CYT-PCM-50, Cytognos, Salamanca, Spain) was used according to manufacturer’s instructions. The Perfect-Count microsphere system contains two different fluorospheres in a known proportion (A and B beads), thereby assuring the accuracy of the assay by verifying the proportion of both types of beads. Known volumes (50 μL) of Perfect-Count microspheres were added to the same known volume (50 μL) of stained blood using a lyse-no-wash technique, with the beads being counted along with cells. Cell viability was measured by staining the samples with the vital dye 7-aminoactinomycin D (7-AAD), as proposed by ISHAGE guidelines[31]. Samples were analysed on a FACSCanto II flow cytometer (BD Biosciences) with a 488-nm argon laser and DIVA 6.1.3 software (BD Bioscience). The gating strategy was in accordance with ISHAGE guidelines[31].
All aspects of the study were approved by our Bellvitge University Hospital’s Research Ethics Committee, and informed consent was obtained from all participants.

Statistical analysis

Data are expressed as mean, median, standard deviation and interquartile range, as appropriate. Continuous variables were compared between groups by means of analysis of variance (ANOVA) or the Mann-Whitney U test, as appropriate. The Wilcoxon signed-rank test and the Friedman test for repeated measures were also used. All tests were performed using SPSS v.13. Statistical significance was set at p < .05.

Results

The initial values of CPC CD34+/μL ranged from 1.54 to 5.41 in the control group (CG) and from 1.83 to 6.88 in the active group (HME). Compared with the CG the HME group showed a very slight increase in the number of CPC on the fourth day after IHH and ME, but this difference was not significant. Figure 1 shows the absolute values of CPC in the HME and CG groups and Figure 2 shows in different graphs individual CPC values for the HEM and CG. There were no statistically significant differences in CPC count either within-subjects (Friedman test) or between groups (Mann-Whitney U test). Multiple comparisons were made by pairwise comparisons of the variables with the Wilcoxon signed-rank test: there was a statistically significant difference between CPC on the second day with respect to pre-intervention CPC (3.38 (IQ range 2.52-6.76) vs 3.35 (IQ range 1.92-5.78,) p = 0.046) and on the fourth day with respect to pre-intervention day in the HME group (4.10 (IQ range 2.51-6.12) cells/uL) vs 3.35 (IQ range 1.92-5.78), p = 0.046). There were no statistically significant differences in the control group; nor were the differences between the HME and the CG significant.
During hypobaric hypoxic periods at a simulated 5000 m altitude, SaO2 and heart rate (HR) were measured five times per session per participant with a pulse oximeter (180 measures). Mean SaO2 was 75.6% (SD 5.6) and mean HR was 89 beats per min (SD 15).

Discussion

This study found no a significant increase in the number of CPC after IHH plus ME, as compared with a control group. This result contrasts with the findings of our previous study, in which IHH plus ME seemed able to increase the concentration of CPC in four healthy men aged around 50 years of age. In this previous experiment the CPC count increased from a median value of 0.95 cells · μL-1 (range: 0.5–2.1) to a median level of 6.65 cells · μL-1 (range: 3.7-10.7), this increase being highly significant[29]. As CPC levels appeared still to be increasing seven days after the last hypoxia session, it was not clear whether a plateau or maximum value had been reached.
The opposite results have been found in some studies using different protocols. Specifically, hypoxia decreased CPC count in two normobaric hypoxia studies involving a total of 20 subjects[32, 33], and also in a hypobaric hypoxia study with 15 mountain trekkers[34]. Different types and intensities of physical exercise have also been shown to be capable of mobilizing CPC. Exercise increased CPC in rats[35] and humans[8, 9, 19, 20, 22, 36], although one study of 20 healthy humans found no such effect[37].
Hypoxia with exercise increased CPC count in three studies[24, 29, 38] and improved haemodynamic and microvascular endothelial function in another[39], although it should be noted that the protocols applied were quite different. Ciulla et al. reported the case of one subject with increased CPC count following the high-altitude hypoxia and exercise oxygen demands of a trek in the Himalayas[38]. As already noted, Viscor et al. reported increased CPC after IHH and ME[29], and Wang et al. found that CPC count increased after five weeks of normobaric hypoxic training on a bicycle ergometer at 60% VO2max[24]. Wang et al applied a different protocol comparing normobaric hypoxia of 15% and 21%, with five days per week of cycling training for five weeks, that is, a total of 25 days of stimulus. On average they found increased CPC, but the responses shown in Figure 2 ranged widely: some subjects presented increases, others decreases, and still others no alteration. For their part, Shatilo et al. showed how ten days of normobaric isocapnic hypoxia improved haemodynamic and microvascular endothelial function in untrained subjects compared with trained healthy senior men[39].
The small samples used in these different studies make it difficult to draw any firm conclusions, since there are many factors that might influence the results. It is worth noting, however, that whereas many types of standard physical exercise can be difficult to implement in the context of hypoxic procedures or when osteoarticular conditions are present, ME is easier to apply and has been shown to be efficient in reproducing training effects[27, 28]. That said, muscle adaptations to electro-stimulation could be different[40], and although normobaric and hypobaric hypoxia are similar, they can provoke different physiological responses[41, 42].
Inter-individual CPC variability[37, 43] and technical variability in CPC enumeration[44] may also influence the measured response. Compared with the CG, the HME group showed a very slight increase in CPC on the fourth day after IHH and ME. The increase was smaller than physiological inter- and intra-individual variations. However, wide variations have been shown in other studies[24, 37, 43, 44]; therefore, we think that it is very important to establish the normal population values, daily spontaneous and/or stimulated physiological changes, the possible different rhythms in healthy people, and the possible variations between responders and non-responders.
Age has been related with less capacity to produce neurogenesis[45] and with different progenitor cell responses[9], although healthy elderly subjects have been shown to be capable of improving CPC clonogenic and migratory capacity after exercise[9]. In this regard, it should be noted that the subjects recruited for the present research were much younger than the four subjects included in our previous study[29]. One possible explanation is that IHH and ME may be more aggressive stimuli in older than for younger subjects. Some studies have reported a time-dependent increase in CPC count after a cycle incremental exercise test under normoxic or hypoxic conditions, increasing 10 min after exercise[21], during 240 min of strenuous exercise[22] and after a marathon race or 1500m field test[19].
Intermittent hypoxia (IH) has been shown to be physiologically beneficial in various groups of healthy and ill people[39, 46, 47]. IH exposure sessions have been used to improve physical condition and to treat several illnesses. Most of these interventions have been derived from studies carried out in the former Soviet Union, although this was done without a clear understanding of the holistic effects[33]. Hypoxia exposure has also been combined with normal athletic training in a variety of ways[48]. More specifically, IHH exposure has been shown to be an efficient stimulus for eliciting different adaptive responses so as to improve peripheral oxygen supply[49, 50]. Serebrovskaya et al. reported similar results with a normobaric hypoxia exposure programme that was able to enhance innate immunity by mobilizing progenitor cells, activating neutrophils and by increasing circulating complement and immunoglobulins[33]. Serebroskaya et al. found an increase in hematopoietic stem and progenitor cells (HSPC) during the final 15 to 20 seconds of a 5-minute normobaric hypoxia bout, which was maintained 15 minutes after four bouts of hypoxia and returned to normal 30 minutes later. After a 14-day program of IHT (intermittent hypoxia treatment), the HSPC count had not changed one day after completion of the program, but had fallen by the seventh day.
The present study does have certain limitations, notably the small sample size and the variability of the techniques used to detect CPC in blood[44]. It is likely that the schedule used for blood sampling also made it difficult to detect rapid and brief transitory changes in CPC, but this study design was based on previous results obtained by our group in which the CPC increase was maintained for seven days. The aim was to find a short stimulus able to maintain the CPC increase over time in order to aid recovery from a variety of diseases. Some studies have reported a time-dependent increase in CPC count after exercise and/or hypoxia[19, 22].

Conclusion

Taking these limitations into account, the results of the present pilot study fail to confirm the significant increase in CPC after IHH plus ME previously reported by our group. It is not possible to know whether the slight increase observed is physiologically relevant, or whether there was also increase at other points after exercise. These results are controversial, and further studies including a larger number of subjects and more exhaustive blood sampling times and technique are required to clarify the possible effect of IHH and ME on the release of progenitor cells. This is particularly important with regard to their possible physiological or pathophysiological significance.

Acknowledgments

This study was supported by DEP2010-22205-C02-02 grant from the Plan Nacional I + D + i 2008-2011 (Spain’s Ministry of Economy and Competitiveness). We would like to thank the volunteers for their participation in the present study.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no conflict of interest to disclose.

Authors’ contributions

All six authors have contributed to the study in accordance with the international consensus on authorship, and they have approved the final draft submitted. LC contributed to the design and implementation of the study, to data collection and analysis, and to manuscript writing. CJ contributed to the conception and design of the study, to its implementation, to data collection and analysis, and to manuscript writing. JB contributed to implementation of the study, performed the measurements of circulating progenitor cells, and contributed to data analysis and manuscript writing. GV contributed to the conception and design of the study, to its implementation, to data collection and analysis, and to manuscript writing. AR contributed to the design and implementation of the study and to manuscript writing. JV made a significant contribution to the conception and design of the study, as well as to data analysis and manuscript writing. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, Finkel T: Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003, 348: 593-600.CrossRefPubMed Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, Finkel T: Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003, 348: 593-600.CrossRefPubMed
2.
Zurück zum Zitat Dimmeler S: Regulation of bone marrow-derived vascular progenitor cell mobilization and maintenance. Arterioscler Thromb Vasc Biol. 2010, 30: 1088-1093.CrossRefPubMed Dimmeler S: Regulation of bone marrow-derived vascular progenitor cell mobilization and maintenance. Arterioscler Thromb Vasc Biol. 2010, 30: 1088-1093.CrossRefPubMed
4.
Zurück zum Zitat Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, Böhm M, Nickenig G: Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005, 353: 999-1007.CrossRefPubMed Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, Böhm M, Nickenig G: Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005, 353: 999-1007.CrossRefPubMed
5.
Zurück zum Zitat Ferrario M, Massa M, Rosti V, Campanelli R, Ferlini M, Marinoni B, De Ferrari GM, Meli V, De Amici M, Repetto A, Verri A, Bramucci E, Tavazzi L: Early haemoglobin-independent increase of plasma erythropoietin levels in patients with acute myocardial infarction. Eur Heart J. 2007, 28: 1805-1813.CrossRefPubMed Ferrario M, Massa M, Rosti V, Campanelli R, Ferlini M, Marinoni B, De Ferrari GM, Meli V, De Amici M, Repetto A, Verri A, Bramucci E, Tavazzi L: Early haemoglobin-independent increase of plasma erythropoietin levels in patients with acute myocardial infarction. Eur Heart J. 2007, 28: 1805-1813.CrossRefPubMed
6.
Zurück zum Zitat Theiss HD, David R, Engelmann MG, Barth A, Schotten K, Naebauer M, Reichart B, Steinbeck G, Franz WM: Circulation of CD34+ progenitor cell populations in patients with idiopathic dilated and ischaemic cardiomyopathy (DCM and ICM). Eur Heart J. 2007, 28: 1258-1264.CrossRefPubMed Theiss HD, David R, Engelmann MG, Barth A, Schotten K, Naebauer M, Reichart B, Steinbeck G, Franz WM: Circulation of CD34+ progenitor cell populations in patients with idiopathic dilated and ischaemic cardiomyopathy (DCM and ICM). Eur Heart J. 2007, 28: 1258-1264.CrossRefPubMed
7.
Zurück zum Zitat Roberts N, Xiao Q, Weir G, Xu Q, Jahangiri M: Endothelial progenitor cells are mobilized after cardiac surgery. Ann Thorac Surg. 2007, 83: 598-605.CrossRefPubMed Roberts N, Xiao Q, Weir G, Xu Q, Jahangiri M: Endothelial progenitor cells are mobilized after cardiac surgery. Ann Thorac Surg. 2007, 83: 598-605.CrossRefPubMed
8.
Zurück zum Zitat Steiner S, Niessner A, Ziegler S, Richter B, Seidinger D, Pleiner J, Penka M, Wolzt M, Huber K, Wojta J, Minar E, Kopp CW: Endurance training increases the number of endothelial progenitor cells in patients with cardiovascular risk and coronary artery disease. Atherosclerosis. 2005, 181: 305-310.CrossRefPubMed Steiner S, Niessner A, Ziegler S, Richter B, Seidinger D, Pleiner J, Penka M, Wolzt M, Huber K, Wojta J, Minar E, Kopp CW: Endurance training increases the number of endothelial progenitor cells in patients with cardiovascular risk and coronary artery disease. Atherosclerosis. 2005, 181: 305-310.CrossRefPubMed
9.
Zurück zum Zitat Hoetzer GL, Van Guilder GP, Irmiger HM, Keith RS, Stauffer BL, DeSouza CA: Aging, exercise, and endothelial progenitor cell clonogenic and migratory capacity in men. J Appl Physiol. 1985, 102: 847-852. 2007CrossRef Hoetzer GL, Van Guilder GP, Irmiger HM, Keith RS, Stauffer BL, DeSouza CA: Aging, exercise, and endothelial progenitor cell clonogenic and migratory capacity in men. J Appl Physiol. 1985, 102: 847-852. 2007CrossRef
10.
Zurück zum Zitat Meng X, Ichim TE, Zhong J, Rogers A, Yin Z, Jackson J, Wang H, Ge W, Bogin V, Chan KW, Thébaud B, Riordan NH: Endometrial regenerative cells: a novel stem cell population. J Transl Med. 2007, 5: 57-PubMedCentralCrossRefPubMed Meng X, Ichim TE, Zhong J, Rogers A, Yin Z, Jackson J, Wang H, Ge W, Bogin V, Chan KW, Thébaud B, Riordan NH: Endometrial regenerative cells: a novel stem cell population. J Transl Med. 2007, 5: 57-PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Kondo T, Hayashi M, Takeshita K, Numaguchi Y, Kobayashi K, Iino S, Inden Y, Murohara T: Smoking cessation rapidly increases circulating progenitor cells in peripheral blood in chronic smokers. Arterioscler Thromb Vasc Biol. 2004, 24: 1442-1447.CrossRefPubMed Kondo T, Hayashi M, Takeshita K, Numaguchi Y, Kobayashi K, Iino S, Inden Y, Murohara T: Smoking cessation rapidly increases circulating progenitor cells in peripheral blood in chronic smokers. Arterioscler Thromb Vasc Biol. 2004, 24: 1442-1447.CrossRefPubMed
12.
Zurück zum Zitat Liu L, Liu H, Jiao J, Liu H, Bergeron A, Dong JF, Zhang J: Changes in circulating human endothelial progenitor cells after brain injury. J Neurotrauma. 2007, 24: 936-943.CrossRefPubMed Liu L, Liu H, Jiao J, Liu H, Bergeron A, Dong JF, Zhang J: Changes in circulating human endothelial progenitor cells after brain injury. J Neurotrauma. 2007, 24: 936-943.CrossRefPubMed
13.
Zurück zum Zitat Flames N, Pla R, Gelman DM, Rubenstein JL, Puelles L, Marin O: Delineation of multiple subpallial progenitor domains by the combinatorial expression of transcriptional codes. J Neurosci. 2007, 27: 9682-9695.CrossRefPubMed Flames N, Pla R, Gelman DM, Rubenstein JL, Puelles L, Marin O: Delineation of multiple subpallial progenitor domains by the combinatorial expression of transcriptional codes. J Neurosci. 2007, 27: 9682-9695.CrossRefPubMed
14.
Zurück zum Zitat Grayson WL, Zhao F, Bunnell B, Ma T: Hypoxia enhances proliferation and tissue formation of human mesenchymal stem cells1. Biochem Biophys Res Commun. 2007, 358: 948-953.CrossRefPubMed Grayson WL, Zhao F, Bunnell B, Ma T: Hypoxia enhances proliferation and tissue formation of human mesenchymal stem cells1. Biochem Biophys Res Commun. 2007, 358: 948-953.CrossRefPubMed
15.
Zurück zum Zitat Zhu LL, Zhao T, Li HS, Zhao H, Wu LY, Ding AS, Fan W, Fan M: Neurogenesis in the adult rat brain after intermittent hypoxia. Brain Res. 2005, 1055: 1-6.CrossRefPubMed Zhu LL, Zhao T, Li HS, Zhao H, Wu LY, Ding AS, Fan W, Fan M: Neurogenesis in the adult rat brain after intermittent hypoxia. Brain Res. 2005, 1055: 1-6.CrossRefPubMed
16.
Zurück zum Zitat Xu Q, Wang S, Jiang X, Zhao Y, Gao M, Zhang Y, Wang X, Tano K, Kanehara M, Zhang W, Ishida T: Hypoxia-induced astrocytes promote the migration of neural progenitor cells via vascular endothelial factor, stem cell factor, stromal-derived factor-1alpha and monocyte chemoattractant protein-1 upregulation in vitro. Clin Exp Pharmacol Physiol. 2007, 34: 624-631.CrossRefPubMed Xu Q, Wang S, Jiang X, Zhao Y, Gao M, Zhang Y, Wang X, Tano K, Kanehara M, Zhang W, Ishida T: Hypoxia-induced astrocytes promote the migration of neural progenitor cells via vascular endothelial factor, stem cell factor, stromal-derived factor-1alpha and monocyte chemoattractant protein-1 upregulation in vitro. Clin Exp Pharmacol Physiol. 2007, 34: 624-631.CrossRefPubMed
17.
Zurück zum Zitat Neubauer JA: Invited review: Physiological and pathophysiological responses to intermittent hypoxia. J Appl Physiol. 1985, 90: 1593-1599. 2001 Neubauer JA: Invited review: Physiological and pathophysiological responses to intermittent hypoxia. J Appl Physiol. 1985, 90: 1593-1599. 2001
18.
Zurück zum Zitat Wilson MH, Newman S, Imray CH: The cerebral effects of ascent to high altitudes. Lancet Neurol. 2009, 8: 175-191.CrossRefPubMed Wilson MH, Newman S, Imray CH: The cerebral effects of ascent to high altitudes. Lancet Neurol. 2009, 8: 175-191.CrossRefPubMed
19.
Zurück zum Zitat Bonsignore MR, Morici G, Riccioni R, Huertas A, Petrucci E, Veca M, Mariani G, Bonanno A, Chimenti L, Gioia M, Palange P, Testa U: Hemopoietic and angiogenetic progenitors in healthy athletes: different responses to endurance and maximal exercise. J Appl Physiol. 2010, 109: 60-67.CrossRefPubMed Bonsignore MR, Morici G, Riccioni R, Huertas A, Petrucci E, Veca M, Mariani G, Bonanno A, Chimenti L, Gioia M, Palange P, Testa U: Hemopoietic and angiogenetic progenitors in healthy athletes: different responses to endurance and maximal exercise. J Appl Physiol. 2010, 109: 60-67.CrossRefPubMed
20.
Zurück zum Zitat Koutroumpi M, Dimopoulos S, Psarra K, Kyprianou T, Nanas S: Circulating endothelial and progenitor cells: Evidence from acute and long-term exercise effects. World J Cardiol. 2012, 4: 312-326.PubMedCentralCrossRefPubMed Koutroumpi M, Dimopoulos S, Psarra K, Kyprianou T, Nanas S: Circulating endothelial and progenitor cells: Evidence from acute and long-term exercise effects. World J Cardiol. 2012, 4: 312-326.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Kroepfl JM, Pekovits K, Stelzer I, Fuchs R, Zelzer S, Hofmann P, Sedlmayr P, Dohr G, Wallner-Liebmann S, Domej W, Mueller W: Exercise increases the frequency of circulating hematopoietic progenitor cells, but reduces hematopoietic colony-forming capacity. Stem Cells Dev. 2012, 21: 2915-2925.CrossRefPubMed Kroepfl JM, Pekovits K, Stelzer I, Fuchs R, Zelzer S, Hofmann P, Sedlmayr P, Dohr G, Wallner-Liebmann S, Domej W, Mueller W: Exercise increases the frequency of circulating hematopoietic progenitor cells, but reduces hematopoietic colony-forming capacity. Stem Cells Dev. 2012, 21: 2915-2925.CrossRefPubMed
22.
Zurück zum Zitat Mobius-Winkler S, Hilberg T, Menzel K, Golla E, Burman A, Schuler G, Adams V: Time-dependent mobilization of circulating progenitor cells during strenuous exercise in healthy individuals. J Appl Physiol. 2009, 107: 1943-1950.CrossRefPubMed Mobius-Winkler S, Hilberg T, Menzel K, Golla E, Burman A, Schuler G, Adams V: Time-dependent mobilization of circulating progenitor cells during strenuous exercise in healthy individuals. J Appl Physiol. 2009, 107: 1943-1950.CrossRefPubMed
23.
Zurück zum Zitat Zaldivar F, Eliakim A, Radom-Aizik S, Leu SY, Cooper DM: The effect of brief exercise on circulating CD34+ stem cells in early and late pubertal boys. Pediatr Res. 2007, 61: 491-495.CrossRefPubMed Zaldivar F, Eliakim A, Radom-Aizik S, Leu SY, Cooper DM: The effect of brief exercise on circulating CD34+ stem cells in early and late pubertal boys. Pediatr Res. 2007, 61: 491-495.CrossRefPubMed
24.
Zurück zum Zitat Wang JS, Lee MY, Lien HY, Weng TP: Hypoxic exercise training improves cardiac/muscular hemodynamics and is associated with modulated circulating progenitor cells in sedentary men. Int J Cardiol. 2014, 170: 315-323.CrossRefPubMed Wang JS, Lee MY, Lien HY, Weng TP: Hypoxic exercise training improves cardiac/muscular hemodynamics and is associated with modulated circulating progenitor cells in sedentary men. Int J Cardiol. 2014, 170: 315-323.CrossRefPubMed
25.
Zurück zum Zitat Ikeda N, Yasu T, Kubo N, Nakamura T, Sugawara Y, Ueda S, Ishikawa SE, Saito M, Kawakami M, Momomura S: Daily exercise and bone marrow-derived CD34+/133+ cells after myocardial infarction treated by bare metal stent implantation. Circ J. 2008, 72: 897-901.CrossRefPubMed Ikeda N, Yasu T, Kubo N, Nakamura T, Sugawara Y, Ueda S, Ishikawa SE, Saito M, Kawakami M, Momomura S: Daily exercise and bone marrow-derived CD34+/133+ cells after myocardial infarction treated by bare metal stent implantation. Circ J. 2008, 72: 897-901.CrossRefPubMed
26.
Zurück zum Zitat Manfredini F, Rigolin GM, Malagoni AM, Catizone L, Mandini S, Sofritti O, Mauro E, Soffritti S, Boari B, Cuneo A, Zamboni P, Manfredini R: Exercise training and endothelial progenitor cells in haemodialysis patients. J Int Med Res. 2009, 37: 534-540.CrossRefPubMed Manfredini F, Rigolin GM, Malagoni AM, Catizone L, Mandini S, Sofritti O, Mauro E, Soffritti S, Boari B, Cuneo A, Zamboni P, Manfredini R: Exercise training and endothelial progenitor cells in haemodialysis patients. J Int Med Res. 2009, 37: 534-540.CrossRefPubMed
27.
Zurück zum Zitat Brocherie F, Babault N, Cometti G, Maffiuletti N, Chatard JC: Electrostimulation Training Effects on the Physical Performance of Ice Hockey Players. Med Sci Sports Exerc. 2005, 37: 455-460.CrossRefPubMed Brocherie F, Babault N, Cometti G, Maffiuletti N, Chatard JC: Electrostimulation Training Effects on the Physical Performance of Ice Hockey Players. Med Sci Sports Exerc. 2005, 37: 455-460.CrossRefPubMed
28.
Zurück zum Zitat Crameri RM, Weston A, Clinstein M, Davis GM, Sutton JR: Effects of electrical stimulation-induced leg training on skeletal muscle adaptability in spinal cord injury. Scand J Med Sci Sports. 2002, 12: 316-322.CrossRefPubMed Crameri RM, Weston A, Clinstein M, Davis GM, Sutton JR: Effects of electrical stimulation-induced leg training on skeletal muscle adaptability in spinal cord injury. Scand J Med Sci Sports. 2002, 12: 316-322.CrossRefPubMed
29.
Zurück zum Zitat Viscor G, Javierre C, Pages T, Ventura JL, Ricart A, Martin-Henao G, Azqueta C, Segura R: Combined intermittent hypoxia and surface muscle electrostimulation as a method to increase peripheral blood progenitor cell concentration. J Transl Med. 2009, 7: 91-PubMedCentralCrossRefPubMed Viscor G, Javierre C, Pages T, Ventura JL, Ricart A, Martin-Henao G, Azqueta C, Segura R: Combined intermittent hypoxia and surface muscle electrostimulation as a method to increase peripheral blood progenitor cell concentration. J Transl Med. 2009, 7: 91-PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Bennie SD, Petrofsky JS, Nisperos J, Tsurudome M, Laymon M: Toward the optimal waveform for electrical stimulation of human muscle. Eur J Appl Physiol. 2002, 88: 13-19.CrossRefPubMed Bennie SD, Petrofsky JS, Nisperos J, Tsurudome M, Laymon M: Toward the optimal waveform for electrical stimulation of human muscle. Eur J Appl Physiol. 2002, 88: 13-19.CrossRefPubMed
31.
Zurück zum Zitat Keeney M, Chin-Yee I, Weir K, Popma J, Nayar R, Sutherland DR: Single platform flow cytometric absolute CD34+ cell counts based on the ISHAGE guidelines. International Society of Hematotherapy and Graft Engineering. Cytometry. 1998, 34: 61-70.CrossRefPubMed Keeney M, Chin-Yee I, Weir K, Popma J, Nayar R, Sutherland DR: Single platform flow cytometric absolute CD34+ cell counts based on the ISHAGE guidelines. International Society of Hematotherapy and Graft Engineering. Cytometry. 1998, 34: 61-70.CrossRefPubMed
32.
Zurück zum Zitat Colombo E, Marconi C, Taddeo A, Cappelletti M, Villa ML, Marzorati M, Porcelli S, Vezzoli A, Bella SD: Fast reduction of peripheral blood endothelial progenitor cells in healthy humans exposed to acute systemic hypoxia. J Physiol. 2012, 590: 519-532.PubMedCentralCrossRefPubMed Colombo E, Marconi C, Taddeo A, Cappelletti M, Villa ML, Marzorati M, Porcelli S, Vezzoli A, Bella SD: Fast reduction of peripheral blood endothelial progenitor cells in healthy humans exposed to acute systemic hypoxia. J Physiol. 2012, 590: 519-532.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Serebrovskaya TV, Nikolsky IS, Nikolska VV, Mallet RT, Ishchuk VA: Intermittent hypoxia mobilizes hematopoietic progenitors and augments cellular and humoral elements of innate immunity in adult men. High Alt Med Biol. 2011, 12: 243-252.PubMedCentralCrossRefPubMed Serebrovskaya TV, Nikolsky IS, Nikolska VV, Mallet RT, Ishchuk VA: Intermittent hypoxia mobilizes hematopoietic progenitors and augments cellular and humoral elements of innate immunity in adult men. High Alt Med Biol. 2011, 12: 243-252.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Mancuso P, Peccatori F, Rocca A, Calleri A, Antoniotti P, Rabascio C, Saronni L, Zorzino L, Sandri MT, Zubani A, Bertolini F: Circulating endothelial cell number and viability are reduced by exposure to high altitude. Endothelium. 2008, 15: 53-58.CrossRefPubMed Mancuso P, Peccatori F, Rocca A, Calleri A, Antoniotti P, Rabascio C, Saronni L, Zorzino L, Sandri MT, Zubani A, Bertolini F: Circulating endothelial cell number and viability are reduced by exposure to high altitude. Endothelium. 2008, 15: 53-58.CrossRefPubMed
35.
Zurück zum Zitat Laufs U, Werner N, Link A, Endres M, Wassmann S, Jurgens K, Miche E, Bóhm M, Nickenig G: Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis. Circulation. 2004, 109: 220-226.CrossRefPubMed Laufs U, Werner N, Link A, Endres M, Wassmann S, Jurgens K, Miche E, Bóhm M, Nickenig G: Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis. Circulation. 2004, 109: 220-226.CrossRefPubMed
36.
Zurück zum Zitat Thijssen DH, Vos JB, Verseyden C, van Zonneveld AJ, Smits P, Sweep FC, Hopman MTE, de Boer HC: Haematopoietic stem cells and endothelial progenitor cells in healthy men: effect of aging and training. Aging Cell. 2006, 5: 495-503.CrossRefPubMed Thijssen DH, Vos JB, Verseyden C, van Zonneveld AJ, Smits P, Sweep FC, Hopman MTE, de Boer HC: Haematopoietic stem cells and endothelial progenitor cells in healthy men: effect of aging and training. Aging Cell. 2006, 5: 495-503.CrossRefPubMed
37.
Zurück zum Zitat Rakobowchuk M, Harris E, Taylor A, Baliga V, Cubbon RM, Rossiter HB, Birch KM: Heavy and moderate interval exercise training alters low-flow-mediated constriction but does not increase circulating progenitor cells in healthy humans. Exp Physiol. 2012, 97: 375-385.PubMedCentralCrossRefPubMed Rakobowchuk M, Harris E, Taylor A, Baliga V, Cubbon RM, Rossiter HB, Birch KM: Heavy and moderate interval exercise training alters low-flow-mediated constriction but does not increase circulating progenitor cells in healthy humans. Exp Physiol. 2012, 97: 375-385.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Ciulla MM, Giorgetti A, Lazzari L, Cortiana M, Silvestris I, Annoni G, De Asmundis C, Fiore AV, Montelatici E, Paliotti R, Magrini F, Rebulla P, Cortelezzi A: High-altitude trekking in the Himalayas increases the activity of circulating endothelial cells. Am J Hematol. 2005, 79: 76-78.CrossRefPubMed Ciulla MM, Giorgetti A, Lazzari L, Cortiana M, Silvestris I, Annoni G, De Asmundis C, Fiore AV, Montelatici E, Paliotti R, Magrini F, Rebulla P, Cortelezzi A: High-altitude trekking in the Himalayas increases the activity of circulating endothelial cells. Am J Hematol. 2005, 79: 76-78.CrossRefPubMed
39.
Zurück zum Zitat Shatilo VB, Korkushko OV, Ischuk VA, Downey HF, Serebrovskaya TV: Effects of intermittent hypoxia training on exercise performance, hemodynamics, and ventilation in healthy senior men. High Alt Med Biol. 2008, 9: 43-52.CrossRefPubMed Shatilo VB, Korkushko OV, Ischuk VA, Downey HF, Serebrovskaya TV: Effects of intermittent hypoxia training on exercise performance, hemodynamics, and ventilation in healthy senior men. High Alt Med Biol. 2008, 9: 43-52.CrossRefPubMed
40.
Zurück zum Zitat Minetto MA, Botter A, Bottinelli O, Miotti D, Bottinelli D, D'Antona G: Variability in Muscle Adaptation to Electrical Stimulation. Int J Sports Med. 2013, 34: 544-553.CrossRefPubMed Minetto MA, Botter A, Bottinelli O, Miotti D, Bottinelli D, D'Antona G: Variability in Muscle Adaptation to Electrical Stimulation. Int J Sports Med. 2013, 34: 544-553.CrossRefPubMed
41.
Zurück zum Zitat Fulco CS, Beidleman BA, Muza SR: Effectiveness of preacclimatization strategies for high-altitude exposure. Exerc Sport Sci Rev. 2013, 41: 55-63.CrossRefPubMed Fulco CS, Beidleman BA, Muza SR: Effectiveness of preacclimatization strategies for high-altitude exposure. Exerc Sport Sci Rev. 2013, 41: 55-63.CrossRefPubMed
42.
Zurück zum Zitat Savourey G, Launay JC, Besnard Y, Guinet-Lebreton A, Alonso A, Sauvet F, Bourrilhon C: Normo or hypobaric hypoxic tests: propositions for the determination of the individual susceptibility to altitude illnesses. Eur J Appl Physiol. 2007, 100: 193-205.CrossRefPubMed Savourey G, Launay JC, Besnard Y, Guinet-Lebreton A, Alonso A, Sauvet F, Bourrilhon C: Normo or hypobaric hypoxic tests: propositions for the determination of the individual susceptibility to altitude illnesses. Eur J Appl Physiol. 2007, 100: 193-205.CrossRefPubMed
43.
Zurück zum Zitat Eidenschink L, DiZerega G, Rodgers K, Bartlett M, Wells DA, Loken MR: Basal Levels of CD34 Positive Cells in Peripheral Blood Differ Between Individuals and are Stable for 18 Months. Cytometry PartB. 2012, 82B: 18-25.CrossRef Eidenschink L, DiZerega G, Rodgers K, Bartlett M, Wells DA, Loken MR: Basal Levels of CD34 Positive Cells in Peripheral Blood Differ Between Individuals and are Stable for 18 Months. Cytometry PartB. 2012, 82B: 18-25.CrossRef
44.
Zurück zum Zitat Lysak D, Kalina T, Martinek J, Pikalova Z, Vokurkova D, Jaresova M, Marinov I, Ondrejkova A, Spacek M, Stehlikova O: Interlaboratory variability of CD34+ stem cell enumeration. A pilot study to national external quality assessment within the Czech Republic. Int J Lab Hematol. 2010, 32: e229-e236.CrossRefPubMed Lysak D, Kalina T, Martinek J, Pikalova Z, Vokurkova D, Jaresova M, Marinov I, Ondrejkova A, Spacek M, Stehlikova O: Interlaboratory variability of CD34+ stem cell enumeration. A pilot study to national external quality assessment within the Czech Republic. Int J Lab Hematol. 2010, 32: e229-e236.CrossRefPubMed
45.
Zurück zum Zitat Elder GA, De GR, Gama Sosa MA: Research update: neurogenesis in adult brain and neuropsychiatric disorders. Mt Sinai J Med. 2006, 73: 931-940.PubMed Elder GA, De GR, Gama Sosa MA: Research update: neurogenesis in adult brain and neuropsychiatric disorders. Mt Sinai J Med. 2006, 73: 931-940.PubMed
46.
Zurück zum Zitat Burtscher M, Pachinger O, Ehrenbourg I, Mitterbauer G, Faulhaber M, Puhringer R, Tkatchouk E: Intermittent hypoxia increases exercise tolerance in elderly men with and without coronary artery disease. Int J Cardiol. 2004, 96: 247-254.CrossRefPubMed Burtscher M, Pachinger O, Ehrenbourg I, Mitterbauer G, Faulhaber M, Puhringer R, Tkatchouk E: Intermittent hypoxia increases exercise tolerance in elderly men with and without coronary artery disease. Int J Cardiol. 2004, 96: 247-254.CrossRefPubMed
47.
Zurück zum Zitat Katayama K, Ishida K, Iwasaki K, Miyamura M: Effect of two durations of short-term intermittent hypoxia on ventilatory chemosensitivity in humans. Eur J Appl Physiol. 2009, 105: 815-821.CrossRefPubMed Katayama K, Ishida K, Iwasaki K, Miyamura M: Effect of two durations of short-term intermittent hypoxia on ventilatory chemosensitivity in humans. Eur J Appl Physiol. 2009, 105: 815-821.CrossRefPubMed
48.
Zurück zum Zitat Levine BD: Intermittent hypoxic training: fact and fancy. High Alt Med Biol. 2002, 3: 177-193.CrossRefPubMed Levine BD: Intermittent hypoxic training: fact and fancy. High Alt Med Biol. 2002, 3: 177-193.CrossRefPubMed
49.
Zurück zum Zitat Esteva S, Pedret R, Fort N, Torrella JR, Pages T, Viscor G: Oxidative stress status in rats after intermittent exposure to hypobaric hypoxia. Wilderness Environ Med. 2010, 21: 325-331.CrossRefPubMed Esteva S, Pedret R, Fort N, Torrella JR, Pages T, Viscor G: Oxidative stress status in rats after intermittent exposure to hypobaric hypoxia. Wilderness Environ Med. 2010, 21: 325-331.CrossRefPubMed
50.
Zurück zum Zitat Panisello P, Torrella JR, Esteva S, Pages T, Viscor G: Capillary supply, fibre types and fibre morphometry in rat tibialis anterior and diaphragm muscles after intermittent exposure to hypobaric hypoxia. Eur J Appl Physiol. 2008, 103: 203-213.CrossRefPubMed Panisello P, Torrella JR, Esteva S, Pages T, Viscor G: Capillary supply, fibre types and fibre morphometry in rat tibialis anterior and diaphragm muscles after intermittent exposure to hypobaric hypoxia. Eur J Appl Physiol. 2008, 103: 203-213.CrossRefPubMed
Metadaten
Titel
Combined intermittent hypobaric hypoxia and muscle electro-stimulation: a method to increase circulating progenitor cell concentration?
verfasst von
Luisa Corral
Casimiro Javierre
Juan Blasi
Ginés Viscor
Antoni Ricart
Josep Lluis Ventura
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2014
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/1479-5876-12-174

Weitere Artikel der Ausgabe 1/2014

Journal of Translational Medicine 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.