Skip to main content
Erschienen in: European Journal of Medical Research 1/2014

Open Access 01.12.2014 | Research

Dehydroepiandrosterone modulates the inflammatory response in a bilateral femoral shaft fracture model

verfasst von: Philipp Lichte, Roman Pfeifer, Britta Elisa Werner, Petra Ewers, Mersedeh Tohidnezhad, Thomas Pufe, Frank Hildebrand, Hans-Christoph Pape, Philipp Kobbe

Erschienen in: European Journal of Medical Research | Ausgabe 1/2014

Abstract

Background

Dehydroepiandrosterone (DHEA) has been shown to have immunomodulatory effects after hemorrhage and sepsis. The present study analyzes whether DHEA is also involved in the mediation of inflammatory stimuli induced by bilateral femoral shaft fracture.

Methods

Male C57/BL6 mice (6 per group) were subjected to closed bilateral femoral shaft fracture with intramedullary nailing followed by administration of either 25 mg/kg/24 h DHEA diluted in saline with 0.1% ethanol or saline with 0.1% ethanol. The sham group was treated by isolated intramedullary nailing without fracture. Animals were sacrificed after 6, 24, or 72 h. Serum TNFα, IL-1β, IL-6, IL-10, MCP-1, and KC concentrations were measured by Bio-Plex ProTm analysis. Acute pulmonary inflammation was assessed by histology, pulmonary myeloperoxidase (MPO) activity, and pulmonary IL-6 concentration.

Results

DHEA was associated with a decrease in the systemic inflammatory response induced by bilateral femoral fracture, especially systemic IL-6 (322.2 vs. 62.5 pg/mL; P = 0.01), IL-1β (1,422.6 vs. 754.1 pg/mL; P = 0.05), and MCP-1 (219.4 vs. 44.1 pg/mL; P >0.01) levels. No changes in pulmonary inflammation were measured.

Conclusion

We conclude that DHEA may be a treatment option to reduce systemic inflammation following musculoskeletal injuries although the pulmonary inflammatory reaction was not affected.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​2047-783X-19-27) contains supplementary material, which is available to authorized users.

Competing interests

All authors declare no conflict of interest.

Authors’ contributions

All authors were involved in the conception of the study. PL, RP, BE, PK, and PE were responsible for animal operations and sample collection. PL, RP, BE, PE, MT, and TP participated in sample analysis. All authors participated in statistical analyses, writing, and editing of the manuscript. All authors read and approved the final manuscript.

Background

Gender differences in susceptibility to complications after hemorrhage and sepsis have been explained by an influence of sex steroids. Dehydroepiandrosterone (DHEA) is a precursor hormone produced in the adrenal glands. It stimulates T-lymphocytes[1] and upregulates the reduced activity of macrophages after trauma which causes a decrease in sepsis-related mortality[2]. Several studies revealed a decreased release of proinflammatory cytokines after DHEA treatment[35]. The effects of DHEA are in part dependent on IL-6[4], but it also exerts IL-6 independent effects. Androstendion, a metabolite of DHEA, improves liver function and perfusion through a reduction of the inducible nitric oxide synthase and endothelin-1 levels after hemorrhage[6]. To our knowledge, the effect of DHEA treatment after musculoskeletal injuries has not been examined.
In previous studies from our group bilateral femoral fractures were associated with a systemic inflammatory response and a migration of inflammatory cells into the pulmonary tissue[79]. These post-traumatic changes might result in a relevant impairment of lung function (for example, acute lung injury (ALI) or Adult Respiratory Distress Syndrome (ARDS)) which represents a frequent complication after long bone fractures in general, but particularly after bilateral femoral shaft fractures[10]. Therefore, the control of systemic inflammation as well as the maintenance of pulmonary function seem to be essential goals in the treatment of musculoskeletal injuries.
In order to further clarify the therapeutic effects of DHEA we tested the following hypothesis: application of DHEA modulates the systemic inflammatory response after bilateral femoral fracture and is associated with changes in organ dysfunction.

Methods

Animals and animal care

The study was approved by the animal welfare committee of the state North Rhine - Westphalia.
During the entire study period all mice were kept by a 12-h light/dark cycle and a constant room temperature. Water and peleted chow were available ad libitum on the ground of the cage. Analgesia was ensured by subcutaneous Buprenorphin 0.1 mg/kg twice a day.

Induction of anesthesia

All procedures were performed under deep anesthesia. A dosage of 50 mg/kg was used as intraperitoneal injection of Phenobarbital. Postoperatively, all mice were placed on warming mats.

Group distribution experimental procedures

Three different groups were included in the experimental design: bilateral femoral shaft fracture with DHEA treatment diluted in saline with 0.1% ethanol (group FxDHEA), bilateral femoral shaft fracture with administration of saline with 0.1% ethanol (group Fx), and Sham group with only intramedullary nailing without fracture (group S). All groups were analyzed at three time points (6, 24, and 72 h) and contained of 18 mice (6 for each time point).

Technique of IM nailing

Femur fractures were induced by a standardized blunt guillotine device as previously described[11]. Retrograde nailing was performed by a small incision lateral to the patella, blunt exposure of the femoral notch, and intramedullary introduction of a 27G needle into the proximal metaphyseal zone. Afterwards the cannula was shortened underneath the cartilaginous surface. The wound was closed by simple interrupted suture.
Six, 24, or 72 h after operation mice were euthanized by exsanguination by cardiac puncture under anesthesia.

Administration of DHEA

Group FxDHEA: DHEA (Sigma-Aldrich, Deisenhofen, Germany) was used in a dosage of 25 mg/kg/24 h. DHEA was dissolved in 70% ethanol. This stock solution was diluted in saline to reach a final ethanol concentration of 0.1%.
Group Fx: Animals of the vehicle group received saline including 0.1% ethanol.
Group S: Application of saline including 0.1% ethanol.
DHEA solution or vehicle solution was injected subcutaneously in the nuchal fold directly after the fracture/sham operation and thereafter every 24 h.

Assessment of TNFα, IL-1β, IL-6, IL-10, MCP-1, and KC plasma concentrations

Time points of measurements were 6, 24, or 72 h. Heparinized blood was centrifuged for 10 min at 5,000 rpm at 10°C. Plasma was separated and stored at -80°C. Concentrations of TNFα, IL-1β, IL-6, IL-10, MCP-1, and KC were measured by Bio-Plex ProTm assays (Biorad, Hercules, CA, USA) according to the manufacturer’s instructions.

Collection of lung samples

The lung was removed under sterile conditions immediately after sacrifice. The right lobe was snap frozen in a microfuge tube. The left lobe was fixed in buffered formalin.

Pulmonary histology

Fixed pulmonary lobes were embedded in paraffin and sliced at 5 μm thickness. Slices were stained with H&E (Hematoxylin and Eosin). Blinded specimen were analyzed by transmitted-light microscopy (Carl Zeiss, Jena, Germany) under 20-fold magnification and the number of inflammatory cells per field of view was counted by two independent examiners.

Assessment of MPO and IL-6 in lung tissue

The frozen lung tissue was thawed and homogenized in a lysis buffer as described by the manufacturer. MPO-enzyme-linked immunosorbent assay kits (MPO ELISA kit, Hycultec GmbH, Beutelsbach, Germany) were used to measure the MPO activity in lung tissue. IL-6 was measured by using standardized ELISA kits (R&D System Inc., Minneapolis, MN, USA). To standardize the MPO and IL-6 levels on the base of the relative protein concentration we used the standardized Pierce® BCA Protein Assay kits (Thermo Scientific, IL, USA). Tissues were diluted 1:4 before measurements.

Statistics

Statistical analyses were performed using SPSS software (SPSS Inc., Chicago, IL, USA). Results are presented as means ± SEM. Data were analyzed by one way analysis of variance and Kruskal-Wallis test. P values below 0.05 were considered statistically significant.

Results

For the study 54 male C57/BL6 mice (Charles River, Germany) aged 8 to 10 weeks with a body weight of 25 ± 2 g were used. Each group (Fx + DHEA, Fx, S) contained 18 mice, six for each of the three time points.

Plasma cytokine response

The systemic release of IL-6 was significantly elevated at 6 h after bilateral femoral shaft fracture compared to the sham group. DHEA treatment was associated with a significantly lower increase of IL-6 levels when compared with group Fx. At 24 h, IL-6 showed a distinct decrease in both fracture groups but was still significantly elevated in the untreated fracture group compared to group Sham (Figure 1A). MCP-1 showed a similar course. It was also markedly increased 6 h after fracture and this increase was significantly suppressed after DHEA administration (Figure 1B).
Serum TNF-α levels were increased at 6 h after fracture and DHEA administration was associated with a decreased systemic concentration (P = 0.065). Plasma levels showed a second peak at 72 h. At this time point DHEA treatment was not associated with any changes in TNF-α levels (Figure 1C). IL-1β showed a similar course as TNF-α: levels were significantly increased after fracture whereby DHEA treatment caused significantly lower serum levels. The second peak at 72 h did not reach significant differences between vehicle and DHEA treatment (Figure 1D).
Systemic levels of KC showed a significant increase after 6 h in both fracture groups. DHEA administration did not result in significant differences compared to the vehicle group (Figure 1E).
IL-10 showed an increase over the time and reached the maximum at 72 h (Figure 1F). No significant effect of DHEA treatment was observed.

Pulmonary inflammation

MPO activity was elevated in mice subjected to bilateral femoral shaft fracture independently of DHEA treatment within the first 24 h (Figure 2A). Both groups demonstrated comparable MPO levels. Pulmonary IL-6 was only significantly elevated at 24 h after bilateral femoral fracture. DHEA administration did not cause significant changes in IL-6 levels (Figure 2B). Furthermore, DHEA treatment did not influence pulmonary infiltration of inflammatory cells following bilateral femoral shaft fracture (Figure 3).

Discussion

Previous studies from our group have shown that long bone fractures induce changes in the systemic and pulmonary inflammatory response. Also, there is an impairment of post-traumatic pulmonary function. In this context, bilateral femur shaft fractures have caused a more sustained inflammatory response than bone injection and unilateral fractures. The systemic and pulmonary inflammatory response after fractures is partly initiated by bone components and mediated via TLR-4 signaling[7]. Furthermore, soft tissue injuries have been shown to be significantly involved in the inflammatory changes after musculoskeletal injuries[12].
Our main results are as follows: DHEA administration was associated with a decrease of systemic inflammatory cytokine levels, especially IL-6, IL-1β, and MCP-1.
In our model the induction of a bilateral femoral fracture was associated by a significant systemic inflammatory response. In accordance to our previous results[8] the main peak of IL-6 and IL-1β levels occurred at 6 h after induction of fractures. The increase of TNFα in our experiments is in contrast to several studies which could not demonstrate an increase after skeletal trauma. This difference might be explained by an enhanced cytokine release due to the fracture associated soft tissue injury in our experimental setting[12]. As previously shown[8], IL-6 release was also markedly stimulated in the early phase after bilateral femoral fracture in this study[8] After 24 h, we observed a significant decrease of IL-6 values. This is in accordance with a decreasing pro-inflammatory response indicated by reduced IL-6, TNFα, and IL-1β levels and an increase of anti-inflammatory cytokine concentrations[13]. The increase of IL-10 levels 72 h after trauma in our experiments may be interpreted as an anti-inflammatory regulatory reaction which causes decreased levels of IL-6 and TNFα.
The DHEA treatment showed a strong and substantial suppression of IL-6 release after 6 h which is in line with the results of other inflammatory models[5]. Also in healthy humans DHEA plasma levels are negatively correlated with systemic IL-6 levels as DHEA inhibits the IL-6 release of mononuclear cells and monocytes[4]. Likewise, in our study IL-1β levels 6 h after fracture were significantly decreased after DHEA administration. The information about effects of DHEA application on the release of IL-1β is sparse. Schmitz et al. observed an increase of systemic IL-1β release after DHEA treatment in septic mice[14]. In contrast, Oberbeck et al. described a depression of TNFα, but no changes in IL-1β levels[15]. However, these results are difficult to interpret, as the current study is the first one to analyze the effects of DHEA after bilateral femoral shaft fracture. This injury pattern might induce alternative physiological mechanisms which might be responsible for the inflammatory reaction[7].
Our study contrasts those investigating effects of hemorrhagic shock and sepsis, as our experiment TNFα release was not significantly depressed after DHEA administration. The strong decrease of TNFα 24 h after fracture might be due to its short half-time period in combination with anti-inflammatory mechanisms. It is well described that TNFα activity is markedly inhibited by autoregulatory processes[3, 16, 17].
The above mentioned increase of IL-10 in the course was not amplified by DHEA treatment.
The activation of neutrophils is among others effected by pro-inflammatory cytokines (for example, IL-8, TNFα, IL-1β, and IL-6), released by macrophages[18]. Whereas the fracture induced increase of some of these cytokines was depressed by DHEA administration KC levels as well as pulmonary inflammation were not affected. KC is known to play an important role in the recruitment of neutrophils during pulmonary inflammation[19]. Therefore the missing influence of DHEA treatment on pulmonary inflammation might be related to unchanged KC levels. Weiss et al. showed that DHEA regulates the inflammatory reaction mainly by a suppression of IL-6 and thereby indirectly inhibits the infiltration of neutrophils[5]. In our model the decrease of pulmonary MPO in the course correlates with the increase of systemic IL-10 which is known to have immunomodulatory effects[16]. The lack of decrease in MPO activity after DHEA treatment correlates with the lack of changes in IL-10 concentrations and the earlier IL-6 suppression.
The lack of effects of DHEA treatment towards pulmonary inflammatory changes may be explained by different injury mechanisms (hemorrhage or sepsis versus bilateral femoral shaft fractures). In this context, pulmonary fat embolism is well known as a major complication after long bone fractures and at least partly responsible for the increased rate of ARDS following bilateral femoral fractures[20, 21]. The unaffected pulmonary inflammatory reaction after DHEA treatment, despite the decreased systemic inflammation in our study, may be explained by the inability of DHEA to influence pulmonary inflammation due to fat embolism. But this hypothesis cannot be proved with the present data.

Drawbacks of the study

The used fracture stabilization does not administer rotational stability which might influence the inflammatory reaction.
Although the effective dosage of DHEA in models of hemorrhage differs in the current literature between 4 and 100 mg/kg/24 h we decided to use a dosage of 25 mg/kg/24 h which has been described in the majority of previous studies in mice. Effects for this dosage have been described especially in sepsis and combined sepsis and trauma models[2224]. Superior effects for higher dosage have not been shown[24].

Conclusion

DHEA treatment was associated with a modulation of the systemic inflammatory response after bilateral femoral fracture. However, the pulmonary response was not affected by the DHEA admission. We conclude that DHEA may be a treatment option to reduce the systemic inflammation following musculoskeletal injuries but organ specific effects have to receive attention.
Open questions concerning the pathogenesis of pulmonary inflammation after long bone fractures and their modulation have to be addressed in further studies.
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/​2.​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

All authors declare no conflict of interest.

Authors’ contributions

All authors were involved in the conception of the study. PL, RP, BE, PK, and PE were responsible for animal operations and sample collection. PL, RP, BE, PE, MT, and TP participated in sample analysis. All authors participated in statistical analyses, writing, and editing of the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Catania RA, Angele MK, Ayala A, Cioffi WG, Bland KI, Chaudry IH: Dehydroepiandrosterone restores immune function following trauma-haemorrhage by a direct effect on T lymphocytes. Cytokine 1999, 11: 443–450. 10.1006/cyto.1998.0458PubMedCrossRef Catania RA, Angele MK, Ayala A, Cioffi WG, Bland KI, Chaudry IH: Dehydroepiandrosterone restores immune function following trauma-haemorrhage by a direct effect on T lymphocytes. Cytokine 1999, 11: 443–450. 10.1006/cyto.1998.0458PubMedCrossRef
2.
Zurück zum Zitat Angele MK, Catania RA, Ayala A, Cioffi WG, Bland KI, Chaudry IH: Dehydroepiandrosterone: an inexpensive steroid hormone that decreases the mortality due to sepsis following trauma-induced hemorrhage. Arch Surg 1998, 133: 1281–1288.PubMedCrossRef Angele MK, Catania RA, Ayala A, Cioffi WG, Bland KI, Chaudry IH: Dehydroepiandrosterone: an inexpensive steroid hormone that decreases the mortality due to sepsis following trauma-induced hemorrhage. Arch Surg 1998, 133: 1281–1288.PubMedCrossRef
3.
Zurück zum Zitat Ben-Nathan D, Padgett DA, Loria RM: Androstenediol and dehydroepiandrosterone protect mice against lethal bacterial infections and lipopolysaccharide toxicity. J Med Microbiol 1999, 48: 425–431. 10.1099/00222615-48-5-425PubMedCrossRef Ben-Nathan D, Padgett DA, Loria RM: Androstenediol and dehydroepiandrosterone protect mice against lethal bacterial infections and lipopolysaccharide toxicity. J Med Microbiol 1999, 48: 425–431. 10.1099/00222615-48-5-425PubMedCrossRef
4.
Zurück zum Zitat Straub RH, Konecna L, Hrach S, Rothe G, Kreutz M, Scholmerich J, Falk W, Lang B: Serum dehydroepiandrosterone (DHEA) and DHEA sulfate are negatively correlated with serum interleukin-6 (IL-6), and DHEA inhibits IL-6 secretion from mononuclear cells in man in vitro: possible link between endocrinosenescence and immunosenescence. J Clin Endocrinol Metab 1998, 83: 2012–2017. 10.1210/jcem.83.6.4876PubMedCrossRef Straub RH, Konecna L, Hrach S, Rothe G, Kreutz M, Scholmerich J, Falk W, Lang B: Serum dehydroepiandrosterone (DHEA) and DHEA sulfate are negatively correlated with serum interleukin-6 (IL-6), and DHEA inhibits IL-6 secretion from mononuclear cells in man in vitro: possible link between endocrinosenescence and immunosenescence. J Clin Endocrinol Metab 1998, 83: 2012–2017. 10.1210/jcem.83.6.4876PubMedCrossRef
5.
Zurück zum Zitat Weiss EP, Villareal DT, Fontana L, Han DH, Holloszy JO: Dehydroepiandrosterone (DHEA) replacement decreases insulin resistance and lowers inflammatory cytokines in aging humans. Aging (Albany NY) 2011, 3: 533–542. Weiss EP, Villareal DT, Fontana L, Han DH, Holloszy JO: Dehydroepiandrosterone (DHEA) replacement decreases insulin resistance and lowers inflammatory cytokines in aging humans. Aging (Albany NY) 2011, 3: 533–542.
6.
Zurück zum Zitat Shimizu T, Szalay L, Choudhry MA, Schwacha MG, Rue LW 3rd, Bland KI, Chaudry IH: Mechanism of salutary effects of androstenediol on hepatic function after trauma-hemorrhage: role of endothelial and inducible nitric oxide synthase. Am J Physiol Gastrointest Liver Physiol 2005, 288: G244-G250. 10.1152/ajpgi.00387.2004PubMedCrossRef Shimizu T, Szalay L, Choudhry MA, Schwacha MG, Rue LW 3rd, Bland KI, Chaudry IH: Mechanism of salutary effects of androstenediol on hepatic function after trauma-hemorrhage: role of endothelial and inducible nitric oxide synthase. Am J Physiol Gastrointest Liver Physiol 2005, 288: G244-G250. 10.1152/ajpgi.00387.2004PubMedCrossRef
7.
Zurück zum Zitat Kobbe P, Kaczorowski DJ, Vodovotz Y, Tzioupis CH, Mollen KP, Billiar TR, Pape HC: Local exposure of bone components to injured soft tissue induces toll-like receptor 4-dependent systemic inflammation with acute lung injury. Shock 2008, 30: 686–691. 10.1097/SHK.0b013e31816f257ePubMedCrossRef Kobbe P, Kaczorowski DJ, Vodovotz Y, Tzioupis CH, Mollen KP, Billiar TR, Pape HC: Local exposure of bone components to injured soft tissue induces toll-like receptor 4-dependent systemic inflammation with acute lung injury. Shock 2008, 30: 686–691. 10.1097/SHK.0b013e31816f257ePubMedCrossRef
8.
Zurück zum Zitat Kobbe P, Vodovotz Y, Kaczorowski DJ, Mollen KP, Billiar TR, Pape HC: Patterns of cytokine release and evolution of remote organ dysfunction after bilateral femur fracture. Shock 2008, 30: 43–47. 10.1097/SHK.0b013e31815d190bPubMedCrossRef Kobbe P, Vodovotz Y, Kaczorowski DJ, Mollen KP, Billiar TR, Pape HC: Patterns of cytokine release and evolution of remote organ dysfunction after bilateral femur fracture. Shock 2008, 30: 43–47. 10.1097/SHK.0b013e31815d190bPubMedCrossRef
9.
Zurück zum Zitat Menzel CL, Pfeifer R, Darwiche SS, Kobbe P, Gill R, Shapiro RA, Loughran P, Vodovotz Y, Scott MJ, Zenati MS, Billiar TR, Pape HC: Models of lower extremity damage in mice: time course of organ damage and immune response. J Surg Res 2011, 166: e149-e156. 10.1016/j.jss.2010.11.914PubMedPubMedCentralCrossRef Menzel CL, Pfeifer R, Darwiche SS, Kobbe P, Gill R, Shapiro RA, Loughran P, Vodovotz Y, Scott MJ, Zenati MS, Billiar TR, Pape HC: Models of lower extremity damage in mice: time course of organ damage and immune response. J Surg Res 2011, 166: e149-e156. 10.1016/j.jss.2010.11.914PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Kobbe P, Micansky F, Lichte P, Sellei RM, Pfeifer R, Dombroski D, Lefering R, Pape HC, TraumaRegister DGU: Increased morbidity and mortality after bilateral femoral shaft fractures: myth or reality in the era of damage control? Injury 2013, 44: 221–225. 10.1016/j.injury.2012.09.011PubMedCrossRef Kobbe P, Micansky F, Lichte P, Sellei RM, Pfeifer R, Dombroski D, Lefering R, Pape HC, TraumaRegister DGU: Increased morbidity and mortality after bilateral femoral shaft fractures: myth or reality in the era of damage control? Injury 2013, 44: 221–225. 10.1016/j.injury.2012.09.011PubMedCrossRef
11.
Zurück zum Zitat Bonnarens F, Einhorn TA: Production of a standard closed fracture in laboratory animal bone. J Orthop Res 1984, 2: 97–101. 10.1002/jor.1100020115PubMedCrossRef Bonnarens F, Einhorn TA: Production of a standard closed fracture in laboratory animal bone. J Orthop Res 1984, 2: 97–101. 10.1002/jor.1100020115PubMedCrossRef
12.
Zurück zum Zitat Kobbe P, Vodovotz Y, Kaczorowski DJ, Billiar TR, Pape HC: The role of fracture-associated soft tissue injury in the induction of systemic inflammation and remote organ dysfunction after bilateral femur fracture. J Orthop Trauma 2008, 22: 385–390. 10.1097/BOT.0b013e318175dd88PubMedCrossRef Kobbe P, Vodovotz Y, Kaczorowski DJ, Billiar TR, Pape HC: The role of fracture-associated soft tissue injury in the induction of systemic inflammation and remote organ dysfunction after bilateral femur fracture. J Orthop Trauma 2008, 22: 385–390. 10.1097/BOT.0b013e318175dd88PubMedCrossRef
13.
Zurück zum Zitat Strecker W, Gebhard F, Rager J, Bruckner UB, Steinbach G, Kinzl L: Early biochemical characterization of soft-tissue trauma and fracture trauma. J Trauma 1999, 47: 358–364. 10.1097/00005373-199908000-00025PubMedCrossRef Strecker W, Gebhard F, Rager J, Bruckner UB, Steinbach G, Kinzl L: Early biochemical characterization of soft-tissue trauma and fracture trauma. J Trauma 1999, 47: 358–364. 10.1097/00005373-199908000-00025PubMedCrossRef
14.
Zurück zum Zitat Schmitz D, Kobbe P, Wegner A, Hammes F, Oberbeck R: Dehydroepiandrosterone during sepsis: does the timing of administration influence the effectiveness. J Surg Res 2010, 163: e73-e77. 10.1016/j.jss.2010.05.017PubMedCrossRef Schmitz D, Kobbe P, Wegner A, Hammes F, Oberbeck R: Dehydroepiandrosterone during sepsis: does the timing of administration influence the effectiveness. J Surg Res 2010, 163: e73-e77. 10.1016/j.jss.2010.05.017PubMedCrossRef
15.
Zurück zum Zitat Oberbeck R, Dahlweid M, Koch R, van Griensven M, Emmendorfer A, Tscherne H, Pape HC: Dehydroepiandrosterone decreases mortality rate and improves cellular immune function during polymicrobial sepsis. Crit Care Med 2001, 29: 380–384. 10.1097/00003246-200102000-00029PubMedCrossRef Oberbeck R, Dahlweid M, Koch R, van Griensven M, Emmendorfer A, Tscherne H, Pape HC: Dehydroepiandrosterone decreases mortality rate and improves cellular immune function during polymicrobial sepsis. Crit Care Med 2001, 29: 380–384. 10.1097/00003246-200102000-00029PubMedCrossRef
16.
Zurück zum Zitat Hildebrand F, Pape HC, Krettek C: The importance of cytokines in the posttraumatic inflammatory reaction. Unfallchirurg 2005, 108: 793–794. 796–803 10.1007/s00113-005-1005-1PubMedCrossRef Hildebrand F, Pape HC, Krettek C: The importance of cytokines in the posttraumatic inflammatory reaction. Unfallchirurg 2005, 108: 793–794. 796–803 10.1007/s00113-005-1005-1PubMedCrossRef
17.
Zurück zum Zitat DeLong WG Jr, Born CT: Cytokines in patients with polytrauma. Clin Orthop Relat Res 2004, 422: 57–65.PubMedCrossRef DeLong WG Jr, Born CT: Cytokines in patients with polytrauma. Clin Orthop Relat Res 2004, 422: 57–65.PubMedCrossRef
18.
Zurück zum Zitat Aldridge AJ: Role of the neutrophil in septic shock and the adult respiratory distress syndrome. Eur J Surg 2002, 168: 204–214. 10.1080/11024150260102807PubMedCrossRef Aldridge AJ: Role of the neutrophil in septic shock and the adult respiratory distress syndrome. Eur J Surg 2002, 168: 204–214. 10.1080/11024150260102807PubMedCrossRef
19.
Zurück zum Zitat Frevert CW, Huang S, Danaee H, Paulauskis JD, Kobzik L: Functional characterization of the rat chemokine KC and its importance in neutrophil recruitment in a rat model of pulmonary inflammation. J Immunol 1995, 154: 335–344.PubMed Frevert CW, Huang S, Danaee H, Paulauskis JD, Kobzik L: Functional characterization of the rat chemokine KC and its importance in neutrophil recruitment in a rat model of pulmonary inflammation. J Immunol 1995, 154: 335–344.PubMed
20.
Zurück zum Zitat Johnson MJ, Lucas GL: Fat embolism syndrome. Orthopedics 1996, 19: 41–48. discussion 48–49PubMed Johnson MJ, Lucas GL: Fat embolism syndrome. Orthopedics 1996, 19: 41–48. discussion 48–49PubMed
22.
Zurück zum Zitat Barkhausen T, Hildebrand F, Krettek C, van Griensven M: DHEA-dependent and organ-specific regulation of TNF-alpha mRNA expression in a murine polymicrobial sepsis and trauma model. Crit Care 2009, 13: R114. 10.1186/cc7963PubMedPubMedCentralCrossRef Barkhausen T, Hildebrand F, Krettek C, van Griensven M: DHEA-dependent and organ-specific regulation of TNF-alpha mRNA expression in a murine polymicrobial sepsis and trauma model. Crit Care 2009, 13: R114. 10.1186/cc7963PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Oberbeck R, Deckert H, Bangen J, Kobbe P, Schmitz D: Dehydroepiandrosterone: a modulator of cellular immunity and heat shock protein 70 production during polymicrobial sepsis. Intensive Care Med 2007, 33: 2207–2213. 10.1007/s00134-007-0851-4PubMedCrossRef Oberbeck R, Deckert H, Bangen J, Kobbe P, Schmitz D: Dehydroepiandrosterone: a modulator of cellular immunity and heat shock protein 70 production during polymicrobial sepsis. Intensive Care Med 2007, 33: 2207–2213. 10.1007/s00134-007-0851-4PubMedCrossRef
24.
Zurück zum Zitat Svec F, Porter JR: The actions of exogenous dehydroepiandrosterone in experimental animals and humans. Proc Soc Exp Biol Med 1998, 218: 174–191. 10.3181/00379727-218-44285PubMedCrossRef Svec F, Porter JR: The actions of exogenous dehydroepiandrosterone in experimental animals and humans. Proc Soc Exp Biol Med 1998, 218: 174–191. 10.3181/00379727-218-44285PubMedCrossRef
Metadaten
Titel
Dehydroepiandrosterone modulates the inflammatory response in a bilateral femoral shaft fracture model
verfasst von
Philipp Lichte
Roman Pfeifer
Britta Elisa Werner
Petra Ewers
Mersedeh Tohidnezhad
Thomas Pufe
Frank Hildebrand
Hans-Christoph Pape
Philipp Kobbe
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2014
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/2047-783X-19-27

Weitere Artikel der Ausgabe 1/2014

European Journal of Medical Research 1/2014 Zur Ausgabe