13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Hypoxia-inducible factors and the prevention of acute organ injury

      review-article
      1 , , 2 , 3
      Critical Care
      BioMed Central

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction Hypoxic preconditioning has long been considered as organ-protective, and its clinical usage has been suggested in elective procedures, such as coronary surgery and organ transplantation. Although the mechanisms have not been clearly elucidated, it has been postulated that changes in cell-membrane composition and upregulation of various cellular protective mechanisms are responsible for a better tolerance of acute injury. Remote preconditioning (i.e., hypoxic stress in one organ conferring resistance to acute hypoxia in other organs) suggests organ cross-talk, perhaps mediated by cytokines and the immune system. Increased expression of heme-oxygenase (HO)-1, heat-shock proteins (HSP), growth factors such as vascular endothelial factor (VEGF), and erythropoietin (EPO) are among the numerous adaptive responses to sublethal injury that are believed to participate in tissue tolerance during subsequent stress. EPO, for instance, is a ubiquitous pleiotropic survival and growth factor that attenuates experimental acute injury in various organ systems, including neuronal, retinal, cardiac, renal, and hepatic tissues. Its clinical efficacy, though suggested in critically ill patients, is yet to be defined [1]. The expression of these protective mediators and many others is regulated by hypoxia-sensing mechanisms through the induction and stabilization of so called hypoxia-inducible factors (HIF) [2]. In this chapter, we will outline the control and action of HIF as key regulators of hypoxic adaptive response, and particularly examine HIF expression during hypoxic stress. We shall discuss recently developed measures that enable HIF signal modification and describe their potential use in conferring tissue tolerance during incipient organ injury. HIF regulation and action HIFs are heterodimers (Figure 1), composed of a constitutive β-subunit (HIF-β) and one of three different oxygen-dependent and transcriptionally active α-subunits, among which HIF-1α and -2α are acknowledged as promotors of hypoxia adaptation, whereas the role of HIF-3α remains unclear. Under normoxia, HIF-α subunits are constantly produced, but not allowed to accumulate, since they are rapidly hydroxylated by oxygen-dependent HIF prolyl-4-hydroxylase domain enzymes (PHD), subsequently captured by the ubiquitin ligase Von-Hippel-Lindau protein (VHL), and degraded by the proteasome. Under oxygen deficiency, PHD activity is reduced, HIF-α accumulates within the cytosol, αβ-dimers are formed, translocate into the nucleus, and bind to hypoxia response elements (HREs) in the promoter enhancer region of genes, which are subsequently transactivated [2-4]. Figure 1 A schematic display of hypoxia-inducible factor (HIF) regulation and biological action. Prolyl-4 hydroxylases (PHDs) serve as oxygen sensors and under normoxic conditions promote degradation of HIF-α isoforms in the proteasome following binding with the ubiquitin ligase, Von-Hippel-Lindau protein (VHL). Hypoxia inhibits PHDs and leads to HIF-α accumulation with HIF-β, and the αβ heterodimer translocates into the nucleus, binds with hypoxia-response elements (HRE) and activates numerous genes important in cell metabolism, proliferation and survival. Many of these genes play a central role in injury tolerance and promotion of tissue oxygenation, such as erythropoietin (EPO), vascular endothelial growth factor (VEGF), inducible NO synthase (iNOS), heme oxygenase (HO)-1, glucose transporter-1, or carbonic anhydrase (CA)-9. Underscored is the inactivation of the HIF-HRE axis by hypoxia, which can be mimicked by carbon monoxide (functional anemia) or by transition metals like cobaltous chloride. Hypoxia-mimetic PHD inhibitors (PHD-I) are potent newly developed measures in the induction of the HIF-HRE axis. For simplicity, numerous additional factors involved in HIF regulation and action are not included in this cartoon and the reader is referred to comprehensive reviews such as references [3,12]. The biological effects of the more than 100 acknowledged HIF target genes are multiple, and include key steps in cell metabolism and survival. Many of the HIF-target genes constitute a reasonable adaptation to hypoxia, such as erythropoiesis (EPO), increased glucose uptake (glucose transporter-1), switch of metabolism to glycolysis (several key enzymes of glycolysis), increased lactate utilization (lactate dehydrogenase), angiogenesis (VEGF), vasodilation (inducible nitric oxide synthase [iNOS]), removal of protons (carbonic anhydrase 9), and scavenging of free radicals (HO-1) [2-4]. Biological and rherapeutic modes of HIF activation Every cell type has the potential to upregulate HIF, principally by the inhibition of PHD, under conditions when cellular oxygen demand exceeds oxygen supply, namely under cellular hypoxia. However, the threshold and extent of HIF activation may depend on the hypoxic stimulus and cell type involved. To some extent, these cellular variations may reflect different expression of various PHD isoforms in different tissues [5-7]. As HIF stimulation may potentiate hypoxia tolerance, studies were conducted to explore its clinical application. Widespread experimental hypoxic stimuli are listed in Table 1, all acting principally by the control of HIF-α degradation, initiated by PHDs. Except for carbon monoxide exposure, which is currently being tested in patients, none of these stimuli seems suitable for preconditional HIF activation in humans. Table 1 Modes of HIF signal enhancement Stimulus/Agent Remarks Potential Clinical Applications Inhibition of PHDs by the induction of cellular physiologic hypoxia  Hypoxic chamber (e.g., 8% O2 in ambient air) depressed systemic PO2  Carbon monoxide admixture to ambient air functional anemia normal systemic PO2 ✓  Anemia normal systemic PO2  Arterial clamping normal systemic PO2 Chemical inhibition of PHDs by hypoxia-mimetics  CoCl2 (interferes with Fe2+) non-specific  Mimosine (2-oxoglutarate analogue) non-specific  Other patented PHD inhibitors specific ✓ Molecular biology techniques  Von-Hippel-Lindau knockout non-specific  PHD siRNA transfection PHD-specific  Constitutively active HIF-α transgenes organ-specific ✓ PHD: prolyl hydroxylase domain enzyme Apart from hypoxic stabilization, widely proven in vivo, HIF activation has also been demonstrated to occur under normal ambient oxygen tensions, mostly in cell cultures challenged with cytokines and growth factors. However, under stress, oxygen demand likely is increased, thus possibly leading to intracellular hypoxia even in cells kept under room air. For technical reasons, it is probably impossible to rule out such local cellular hypoxia that may exist predominantly within the mitochondria. Beyond this academic distinction between true cellular hypoxia and normoxia, it is important to recognize that clinical conditions, like inflammation, infection and sepsis, may lead to HIF activation. Thus, theoretically, cytokines or growth factors could be used for preconditional HIF activation in humans. Although not a reasonable therapeutic intervention, strong and stable normoxic HIF activation can be achieved by deletion of the VHL gene, which is a constant phenomenon in Von Hippel Lindau Disease and in renal clear cell carcinoma, and is also encountered in other tumors. Transgenic animals with VHL knockout serve to test the potential of HIF activation in ischemic/hypoxic diseases (C Rosenberger, unpublished data) [8]. Additional experimental probes for enhancing HIF signal are by transfection with PHD siRNA [9] or with the generation of constitutively active HIF-α transgenes [10]. So-called hypoxia mimetics block PHD activity, thus upregulating HIF under normoxia. PHDs require 2-oxoglutarate and ferrous iron as co-substrates. Non-specific PHD inhibitors are either 2-oxoglutarate analogues or interfere with Fe2+. Recently, more specific PHD inhibitors (PHD) have been synthesized [11], and are currently being tested in animal and human studies. Figure 1 represents a simplistic scheme of the canonical HIF regulation and action. Recent discoveries underscore a host of additional compound biological pathways, associated with the regulation of the HIF signal, including the control of HIF synthesis, HIF controlling PHD synthesis, putative competing/intervening impacts of HIF-3α and PHD-3, cross-talk of HIF and other key regulators of gene expression (STAT, p-300 and others), further modification of HIF-α activity at the level of DNA hypoxia-responsive elements by small ubiquitin-like modifiers (SUMO) and factor inhibiting HIF (FIH), and the effect of reactive oxygen species (ROS), NO and Krebs cycle metabolites on HIF degradation. These complex pathways are beyond the scope of this review, and the interested reader is referred to additional references [3,5,12-18]. HIF expression under hypoxic stress and tissue injury The kidney serves as an excellent example for under-standing HIF expression under hypoxic stress. Renal oxygenation is very heterogeneous, with PO2 falling to levels as low as 25 mmHg in the outer medulla under normal physiologic conditions and to even lower values in the papilla [3,4,19]. Changes in renal parenchymal microcirculation and oxygenation have been thoroughly investigated in acute and chronic renal disorders [19,20]. Finally, the complex renal anatomy in which different cell types are in close proximity to regions with comparable ambient oxygenation, enables comparisons of cellular HIF response. Interestingly, HIF expression is below detection thresh-old by immunostaining in the renal medulla, despite low physiologic ambient oxygenation (It should be emphasized that this statement regarding negative HIF immunostaining in the normally hypoxic medulla relates to kidneys perfusion-fixed in vivo without an interruption of renal oxygenation before fixation. Other modes of tissue harvesting for HIF determination, either by immunostaining or by molecular biology techniques may be falsely positive, as hypoxia-induced inhibition of PHD activity is instantaneous, and may lead to HIF-α stabilization even over short periods of hypoxia). Conceivably, this reflects the plasticity of HIF control to adjust for 'physiologically normal' oxygenation (i.e., adjusted rates of HIF-α generation and degradation under normal conditions. Enhanced renal HIF-α is noted in rodents subjected to hypoxia or to inhaled carbon monoxide (chemical hypoxia) [21], and in hypoxic isolated perfused kidneys [22]. Different cells express diverse HIF isoforms: Whereas tubular segments express HIF-1α, HIF-2α is principally produced by vascular endothelial and interstitial cells [21-23]. Interestingly, HIF-dependent genes are also selectively expressed in different cell types. For instance HIF-2-triggered EPO generation is specifically found in interstitial cells in the deep cortex [24]. In hypoxic isolated perfused kidneys, attenuation of severe medullary hypoxia by the inhibition of tubular transport markedly enhanced HIF expression, probably under-scoring a window of opportunity to generate HIF and HIF-mediated adaptive responses only under moderate and sublethal hypoxic stress [22]. This pattern is consistent with HIF expression at the border of renal infarct zones only, indicating that dying cells within the critically ischemic region are incapable of mounting a hypoxia adaptive response [25]. We also found that HIF-α isoforms are stabilized in acute hypoxic stress, predominantly in the cortex in rhabdomyolysis-induced kidney injury [26], in the outer stripe of the outer medulla following ischemia and reperfusion [27,28], or in the inner stripe and inner medulla following the induction of distal tubular hypoxic injury by radiocontrast agent, or after the inhibition of prostaglandin or NO synthesis or with their combinations [23]. Outer medullary HIF stabilization is also noted in chronic tubulointerstitial disease [29] and in experimental diabetes [30], again spatially distributed in areas with proven hypoxia. HIF was also detected in biopsies from transplanted kidneys [31]. Thus, HIF immunostaining is chronologically and spatially distributed in renal regions with abnormally low PO2. Normal mice subjected to warm ischemia and reperfusion display limited injury only, as compared with extensive damage in HIF (+/-) mice [32]. Thus, the importance of mounting an HIF response during hypoxic stress is undeniable. Hypoxia-driven HIF stabilization during hypoxic stress has been encountered in other organs as well. HIF-1α and PHD-2 expression increased in the neonatal rat brain following hypoxia [33] and HIF was detected in the hypoxic subendocardium [34] and in the ischemic liver [27]. HIF is also found within hypoxic regions in tumors, and may play an important role in tumor progression via upregulation of growth promoting and angiogenic factors [35]. Potential usage of HIF modulation in clinical practice The impact of HIF stimulation on the expression of HIF-dependent tissue-protective genes led to the expectation that timely upstream HIF stimulation may have great potential in the protection of endangered organs by downstream induction of protective genes [12]. Indeed, repeated systemic hypoxia, for instance, results in enhanced expression of renal HIF and HIF-dependent genes and attenuates warm-ischemic injury [36]. The use of hypoxia-mimetic PHD inhibitors is a promising potential new treatment option in diseases such as myocardial infarction, stroke, renal or liver injury, peripheral vascular disease, or severe anemia. Studies with PHD inhibitors and other manipulations of HIF upregulation favor this hypothesis [11]. Anemia Specific PHD inhibitors induce HIF-2α expression in interstitial fibroblasts in the deep cortex [24], enhance erythropoietin generation, and were found to provoke erythrocytosis in primates [37]. Phase 2 clinical trials in patients with chronic kidney disease are currently under way, studying the effect of oral PHD inhibitors as potential substitutes to EPO injection. Acute kidney injury The potential protective impact of HIF upregulation by PHD inhibitors has been extensively studied in acute kidney injury. In isolated kidneys perfused with low-oxygen containing medium, pre-treatment with a PHD inhibitor improved renal blood flow and attenuated medullary hypoxic damage [38]. Conditional inactivation of VHL in mice (hence HIF stabilization) resulted in tolerance to renal ischemia and reperfusion [8] and to rhabdomyolysis-induced acute kidney injury (Rosen-berger C, unpublished data). Whereas gene transfer of negative-dominant HIF led to severe damage in the normally hypoxic renal medulla in intact rats, transfer of constitutively active HIF (HIF/VP16) induced expression of various HIF-regulated genes and protected the medulla against acute ischemic insults [39]. Furthermore, in rats and mice subjected to warm ischemia and reflow, PHD inhibitors and carbon monoxide pre-treatment (i.e., functional anemia) markedly attenuated kidney damage and dysfunction [32,40]. Donor pre-treatment with a PHD inhibitor also prevented graft injury and prolonged survival in an allogenic kidney transplant model in rats [41]. Finally, rats preconditioned by carbon monoxide, displayed reduced cisplatin renal toxicity, with attenuation of renal dysfunction and the extent of tubular apoptosis and necrosis [42]. Taken together, all these observations indicate that HIF stabilization seemingly is a promising novel interventional strategy in acute kidney injuries [12]. Myocardial injury Activation of the HIF system has also been found to be cardioprotective. In a model of myocardial ischemia in rabbits, pre-treatment with a PHD inhibitor induced robust expression of HO-1 and markedly attenuated infarct size and myocardial inflammation [43]. In another report, PHD inhibitors did not reduce infarct size, but improved left ventricular function and prevented remodeling [44]. In the same fashion, selective silencing of PHD-2 with siRNA 24 h before global myocardial ischemia/reperfusion in mice reduced the infarct size by 70% and markedly improved left ventricular systolic function [9]. Remote preconditioning by intermittent renal artery occlusion also resulted in cardiac protection, conceivably through PHD inhibition [45]. Enhanced levels of PHD-3 were traced in the hibernating myocardium [34] and in end-stage heart failure in humans, associated also with elevated HIF-3α [46] (which may act as a competitive inhibitor of active HIF-α isoforms [14]). Thus, PHD inhibitors may conceivably also be beneficial in these disorders. Finally, cardioprotection during heat acclimation is also mediated in part by HIF upregulation [47], providing another potential situation for the administration of PHD inhibitors. Neuronal injuries The effect of PHD inhibitors has also been assessed in disorders of the central nervous system. In vitro, rotenone-induced neuronal apoptosis was attenuated and autophagy increased, as the result of enhanced HIF following deferoxamine administration [48]. In vivo, PHD inhibitors have shown promising results in the attenuation of ischemic stroke [49], and might be neuroprotective in metabolic chronic neurodegenerative conditions [50]. However, studies showing inhibition of PHD-1 by ROS suggest non-HIF-mediated neuronal protection under normoxic conditions [51]. Lung injury Preterm lambs developing respiratory distress syndrome display upregulation of PHDs with a reciprocal fall in HIF-α isoforms and HIF-dependent VEGF [53]. This observation implies that PHD inhibitors might have therapeutic potential in this clinical setup. Liver disease Hepatic HIF-1α is upregulated following warm ischemia [27], and is required for restoration of gluconeogenesis in the regenerating liver [52], implying yet another potential use for PHD inhibitors in acute liver disease. Peripheral vascular disease In a model of limb ischemia in mice, PHD inhibitors enhanced HIF expression and downstream VEGF and VEGF-receptor Flk-1, leading to improved capillary density, indicating a potential therapeutic use of PHD inhibitors in promoting angiogenesis in ischemic diseases, such as severe peripheral vascular disease [54]. Transfection with HIF-1α, combined with PHD inhibitor-treated bone marrow-derived angiogenic cells increased perfusion, motor function, and limb salvage in old mice with ischemic hind limbs [55]. Results of a phase-1 study in patients with critical limb ischemia indicate that transfection with a constitutively active form of HIF-1α might also promote limb salvage [10]. Further clinical trials with PHD inhibitors are currently under way in burn wound healing and salvage of critically ischemic limbs. Oxidative stress Enhanced cellular ROS concentrations, as happens with shock and tissue hypoxia, result in increased PHD activity, and this effect is antagonized by ROS scavengers [15]. This situation may lead to HIF de-stabilization and inadequate HIF response to hypoxia. For example, hypoxia-mediated HIF expression in the diabetic renal medulla is substantially improved by the administration of the membrane-permeable superoxide dismutase mimetic tempol [30]. It is, therefore, tempting to assume that ROS scavengers, as well as PHD inhibitors may improve tissue adaptive responses to hypoxia, coupled with oxidative stress. However, contradicting evidence exists, indicating that ROS might trigger HIF in the absence of hypoxia. This has been suggested by studying liver tissue in acetaminophen-induced liver injury, before the development of overt liver injury and hypoxia [56], and in aged well-fed animals [57]. The role of HIF stimulation during oxidative stress therefore needs further assessment. Important considerations HIF stimulation is not all-protective. The wide range of HIF-dependent genes, and its tight cross-communication with other key regulators of gene expression [13,58,59] raise concern regarding concomitant non-selective activation of protective as well as harmful systems. Among potential unwanted outcomes is the enhancement of tumor growth [60], promotion of fibrosis [61] or the induction of pre-eclampsia in pregnant women [62]. Indeed, whereas HIF activation is considered renoprotective in acute kidney injury, it may play a role in the progression of chronic kidney disease and certainly is an important factor in the promotion of renal malignancy [3,20]. Diverse characteristics and distribution patterns of different PHDs [5-7] and particular actions of various PHD inhibitors [11,37] might enable selective manipulation of the HIF system in a more desired way, selectively favoring advantageous HIF-dependent responses in preferred tissues. Furthermore, it is believed that activation of adverse responses requires protracted HIF stimulation, whereas short-term and transient HIF activation might suffice to activate tissue-protective systems without continuing induction of harmful systems. However, this concept needs confirmation in clinical trials. Conclusion Elucidating the mechanisms involved in HIF-mediated cellular responses to acute hypoxic stress has led to the discovery of novel potential therapeutic options for the prevention or attenuation of tissue injury. The non-selective enhancement of gene expression by current modes of HIF augmentation warrants caution, since undesired enhancement of certain genes may be hazardous. We anticipate that in the coming years the use of PHD inhibitors and other stimulants of the HIF system will be tested in many clinical scenarios associated with critical care and emergency medicine, while HIF silencing strategies may be tested in chronic diseases, such as malignancies and disorders with enhanced tissue scarring. Competing interests The authors declare that they have no competing interests. List of abbreviations used EPO: erythropoietin; FIH: factor inhibiting HIF; HIF: hypoxia-inducible factors; HO: heme-oxygenase; HRE: hypoxia response elements; HSP: heat-shock proteins; PHD: prolyl-4-hyrdoxylase domain enzymes; ROS: reactive oxygen species; SUMO: small ubiquitin-like modifiers; VEGF: vascular endothelial growth factor; VHL: Von-Hippel-Lindau protein.

          Related collections

          Most cited references54

          • Record: found
          • Abstract: found
          • Article: not found

          Signal transduction to hypoxia-inducible factor 1.

          Hypoxia-inducible factor 1 (HIF-1) is a transcriptional activator that functions as a master regulator of O2 homeostasis. HIF-1 target genes encode proteins that increase O2 delivery and mediate adaptive responses to O2 deprivation. HIF-1 activity is regulated by the cellular O2 concentration and by the major growth factor-stimulated signal transduction pathways. In human cancer cells, both intratumoral hypoxia and genetic alterations affecting signal transduction pathways lead to increased HIF-1 activity, which promotes angiogenesis, metabolic adaptation, and other critical aspects of tumor progression.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Inhibition of hypoxia inducible factor hydroxylases protects against renal ischemia-reperfusion injury.

            Acute renal failure resulting from hypoperfusion and hypoxia is a significant clinical problem. Hypoxia activates the heterodimeric transcription factor hypoxia inducible factor (HIF), leading to changes in gene expression that promote tissue adaptation and survival. To determine whether HIF may protect the kidney from ischemia-reperfusion injury, we subjected hif1a(+/-) and hif2a(+/-) mice to renal ischemia-reperfusion injury. Injury was substantially more severe in hif(+/-) than in littermate controls, consistent with a protective role for HIF. Because wild-type mice exhibited submaximal HIF accumulation in response to no-flow ischemia, we tested compounds that might augment the protective HIF response following ischemia-reperfusion in these animals. We found that l-mimosine and dimethyloxalylglycine, two small molecules that activate HIF by inhibiting HIF hydroxylases, protected mouse kidneys from ischemia-reperfusion injury. Therefore, pharmacological activation of HIF may offer an effective strategy to protect the kidney from ischemic injury.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Expression of hypoxia-inducible factor-1alpha and -2alpha in hypoxic and ischemic rat kidneys.

              Oxygen tensions in the kidney are heterogeneous, and their changes presumably play an important role in renal physiologic and pathophysiologic processes. A family of hypoxia-inducible transcription factors (HIF) have been identified as mediators of transcriptional responses to hypoxia, which include the regulation of erythropoietin, metabolic adaptation, vascular tone, and neoangiogenesis. In vitro, the oxygen-regulated subunits HIF-1alpha and -2alpha are expressed in inverse relationship to oxygen tensions in every cell line investigated to date. The characteristics and functional significance of the HIF response in vivo are largely unknown. High-amplification immunohistochemical analyses were used to study the expression of HIF-1alpha and -2alpha in kidneys of rats exposed to systemic hypoxia bleeding anemia, functional anemia (0.1% carbon monoxide), renal ischemia, or cobaltous chloride (which is known to mimic hypoxia). These treatments led to marked nuclear accumulation of HIF-1alpha and -2alpha in different renal cell populations. HIF-1alpha was mainly induced in tubular cells, including proximal segments with exposure to anemia/carbon monoxide, in distal segments with cobaltous chloride treatment, and in connecting tubules and collecting ducts with all stimuli. Staining for HIF-1alpha colocalized with inducible expression of the target genes heme oxygenase-1 and glucose transporter-1. HIF-2alpha was not expressed in tubular cells but was expressed in endothelial cells of a small subset of glomeruli and in peritubular endothelial cells and fibroblasts. The kidney demonstrates a marked potential for upregulation of HIF, but accumulation of HIF-1alpha and HIF-2alpha is selective with respect to cell type, kidney zone, and experimental conditions, with the expression patterns partly matching known oxygen profiles. The expression of HIF-2alpha in peritubular fibroblasts suggests a role in erythropoietin regulation.
                Bookmark

                Author and article information

                Journal
                Crit Care
                Critical Care
                BioMed Central
                1364-8535
                1466-609X
                2011
                22 March 2011
                22 March 2012
                : 15
                : 2
                : 209
                Affiliations
                [1 ]Department of Medicine, Hadassah Hosptial, Mt. Scopus, PO Box 24035, 91240 Jerusalem, Israel
                [2 ]Department of Pathology, Beth Israel Deaconess Medical Center and Harvard University, 330 Brookline Avenue, Boston, MA 02215, USA
                [3 ]Department of Trauma/General Surgery, UPMC - Presbyterian Hospital, F1266 Lothrop Street, Pittsburgh, PA, 15213, USA
                Article
                cc9991
                10.1186/cc9991
                3219405
                21457510
                0ca65e65-fbeb-4adb-b7fa-7ad85e4b797f
                Copyright ©2011 Springer-Verlag Berlin Heidelberg.

                This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilm or in any other way, and storage in data banks. Duplication of this publication or parts thereof is permitted only under the provisions of the German Copyright Law of September 9, 1965, in its current version, and permission for use must always be obtained from Springer-Verlag. Violations are liable for prosecution under the German Copyright Law.

                History
                Categories
                Review

                Emergency medicine & Trauma
                Emergency medicine & Trauma

                Comments

                Comment on this article