Skip to main content
Erschienen in: Journal of Cachexia, Sarcopenia and Muscle 3/2011

Open Access 01.09.2011 | Review

The role of myostatin in muscle wasting: an overview

verfasst von: Yulia Elkina, Stephan von Haehling, Stefan D. Anker, Jochen Springer

Erschienen in: Journal of Cachexia, Sarcopenia and Muscle | Ausgabe 3/2011

Abstract

Myostatin is an extracellular cytokine mostly expressed in skeletal muscles and known to play a crucial role in the negative regulation of muscle mass. Upon the binding to activin type IIB receptor, myostatin can initiate several different signalling cascades resulting in the upregulation of the atrogenes and downregulation of the important for myogenesis genes. Muscle size is regulated via a complex interplay of myostatin signalling with the insulin-like growth factor 1/phosphatidylinositol 3-kinase/Akt pathway responsible for increase in protein synthesis in muscle. Therefore, the regulation of muscle weight is a process in which myostatin plays a central role but the mechanism of its action and signalling cascades are not fully understood. Myostatin upregulation was observed in the pathogenesis of muscle wasting during cachexia associated with different diseases (i.e. cancer, heart failure, HIV). Characterisation of myostatin signalling is therefore a perspective direction in the treatment development for cachexia. The current review covers the present knowledge about myostatin signalling pathways leading to muscle wasting and the state of therapy approaches via the regulation of myostatin and/or its downstream targets in cachexia.

1 Introduction

Cachexia is a syndrome occurring at terminal stages of diseases such as cancer, chronic heart failure, chronic kidney failure or AIDS [1]. This syndrome is characterized by loss of body weight as a consequence of pathological changes in different metabolic pathways. It leads to increased morbidity and mortality irrespective of the underlying disease. A major role in the development of cachexia is played by the loss of muscle mass accompanied by the loss of fat [2]. Such muscle hypotrophy is the result of multiple alterations at the molecular level, e.g. the disturbance in the balance between protein degradation and protein synthesis [1]. Proinflammatory cytokines such as interleukin-1, interleukin-6 and tumour necrosis factor-α (TNFα) were shown to play an important role in the development of muscle wasting [3]. However, there is no established treatment for cachexia based on the regulation of their signalling. Many different peptides have received therapeutic interest over the last decade, including ghrelin, leptin, melanocortins and growth hormone [46]. In 1997, the role of another extracellular factor in the negative regulation of muscle mass, referred to as myostatin, was discovered [7]. Myostatin upregulation was found in the pathogenesis of cancer, HIV, heart failure associated cachexia and aging [812]. It has become one of the main targets in the investigation of the regulation of muscle mass.

2 Myostatin

Myostatin, also known as growth/differentiation factor-8 (GDF-8) is a member of tumour growth factor β (TGF-β) family [7]. This protein is a homodimer with a molecular weight of 25 kDa and a disulfide bond between the monomers at the C-terminal regions [7]. Myostatin circulates in the blood in a latent form with an additional non-covalently bound propeptide at the N-terminus. Proteolytic cleavage of the propeptide by the bone morphogenetic protein (BMP)-1/tolloid family of metalloproteinases is necessary for activation of protein function [13].
The role of myostatin in skeletal muscle was discovered using the method of gene disruption in mice. Mstn null animals showed significant increase in muscle mass (up to two-fold) and decrease of fat tissue compared to the wild type [14, 15]. Similar effects were observed in the presence of natural mutations of Mstn in cattle, sheep, dogs and humans [1619] and upon the inhibition of the protein function in adult mice [20]. At the same time, overexpression of Mstn led to the reduction of muscle mass suggesting myostatin to be a negative regulator of skeletal muscle growth. During embryogenesis, myostatin is exclusively expressed in skeletal muscle [7] to control the differentiation and proliferation of the myoblast, but in adulthood, it is not only restricted to skeletal muscle but also detected in other tissues (e.g. heart, adipose tissue, mammary gland) [7, 2125].
The expression of myostatin in the healthy heart is low and mostly detected in Purkinje fibres but not in cardiomyocytes [22]. After myocardial infarction, increased expression of myostatin was observed in sheep, but its expression was restricted to the damaged zone [22]. Moreover, George et al. recently published new data suggesting the increase of myostatin levels in the failing heart [26]. The authors also showed an increase of its latent complex in circulation and expression of BMP-1 that could explain the development of cachexia in patients with heart failure [26]. These data are in accordance with the recent research of Heineke et al. who suggested that myostatin produced by cardiomyocytes could stimulate muscle wasting in heart failure [27].
The role of myostatin in the regulation of adipose tissue is not well understood. Myostatin and its receptor (type IIB activin receptor, ActRIIB) were shown to be expressed in adipose tissue at low levels [7, 14]. Several studies showed the negative effect of myostatin on preadipocyte differentiation and proliferation [28, 29]. Contrary, an effect of promotion of the differentiation leading to the increased adipogenesis was observed in pluripotent mesenchymal cells [30, 31]. Moreover, Mstn null mice had a decreased amount of adipose tissue [14, 32]. It is still unclear whether the effect of myostatin on adipose tissue is the direct result of regulation or it is an indirect consequence of skeletal muscle growth. Reduced adiposity was observed in different transgenic mouse models of muscle hypertrophy [3335]. Thereby the decreased amount of adipose tissue in Mstn null mice could be an indirect drawback of significant increases in muscle mass. Such an effect can be mediated through leptin—adipose-specific hormone regulating food intake and energy homeostasis. Its level was shown to be lower in Mstn null mice [32]. Therefore, additional experiments are required to determine the role of myostatin in adipose tissue.

3 Binding to the cell

Myostatin is an extracellular cytokine, and as many other members of the TGF-β family, it mediates the signal through activin receptors [36]. Active myostatin mostly binds to the ActRIIB [36] and engages the signalling cascade leading to the inhibition of myoblast differentiation and proliferation (Fig. 1). ActRIIB can mediate other signalling pathways with diverse affinity for ligands—high for activin A and GDF11, low for BMP-2 and BMP-7 [38]. These ligands are responsible for several cell responses by activating different members of the Smad family of transcription factors.
Activin receptors are transmembrane threonine/serine kinases divided in two types. Type I receptor (ALK 4 and ALK 5 for myostatin [28]) has the unique GS domain located closely to the intracellular space and adjacent to the kinase domain which is absent in the second type (ActRII). Binding of myostatin with ActRIIB causes its assembly with type I receptor and phosphorylation of its GS domain. Therefore, the signal of myostatin is mediated through activated complex of two receptors [39].
Myostatin signalling through ActRIIB is crucial for the regulation of muscle growth. The natural defect in ActRIIB sensitivity in humans leads to a significant increase in muscle mass [40]. The same effect was observed in the experiments that used blockade of the murine receptor [40]. In the characterization of the ActRIIB role in myostatin signalling, the soluble form of this receptor (sActRIIB) is used. sActRIIB is a fusion protein of the receptor extracellular domain with immunoglobulin Fc. It acts as a decoy receptor for myostatin. Healthy mice treated with sActRIIB showed a 60% increase in muscle mass, just 2 weeks after treatment initiation [40]. Surprisingly, in mice treated with antimyostatin antibodies (JA-16) or myostatin propeptide known to bind circulating myostatin, the hypertrophy of muscle was lower compared to sActRIIB [41, 42]. Moreover, in Mstn knockout mice treated with the sActRIIB, an increase in muscle mass by 15–25% was observed [40]. This suggests the presence of at least one more ligand that binds to the receptor thereby regulating the mass of skeletal muscle. Souza et al. proposed that such ligands could embrace BMP-11, activin A, B and AB, but this hypothesis requires more evidence [43]. However, it could explain why myostatin blockade in clinical trials had no significant effect on muscle weight [44]. The results obtained in cancer models of Lewis lung carcinoma and B16F10 melanoma are in agreement with this theory. It was shown that myostatin knockout mice had a more pronounced tendency to develop muscle wasting than wild-type animals. This finding suggests that myostatin is not the only ligand providing the signal leading to muscle wasting. On the other hand, the same study showed that myostatin could prevent muscle wasting to a certain degree [45].
Recently, Zhou et al. moved a significant step forward in development of a novel therapy [46]. The authors demonstrated that sActRIIB prevents or even reverses development of cancer cachexia. Their research showed the connection between the development of cancer cachexia and the activation of activin receptor. Blockade of ActRIIB led to the regeneration of muscle and cardiac mass [46]. These data suggest a fundamental role of ActRIIB-mediated signalling pathways in the induction of muscle wasting during cachexia and can be used in the development of treatments against cachexia.

4 Myostatin extracellular regulation

Myostatin circulates in the blood in a latent complex with non-covalently bound propeptide at the N-terminus [13, 36, 47], crucial for the correct folding of the protein [20]. The propeptide was shown to bind the active myostatin in vivo and in vitro, and its overexpression in mice results in an increase of muscle mass [36, 48].
Specific inhibitors can prevent binding of myostatin to ActRIIB in serum. One of them is follistatin, an extracellular cysteine-rich glycoprotein with a structure not similar to members of TGF-β family. Follistatin binds myostatin and inhibits its activity by preventing its binding to the receptor [11, 36]. Overexpression of follistatin in vivo leads to muscle hypertrophy similar to the one observed in Mstn null mice [36]. Mice with homozygous mutation in the Fst gene showed a decrease in muscle mass suggesting an important role of follistatin in the regulation of myogenesis [49]. Follistatin-like 3 protein has more than 30% homology with follistatin and was also shown to bind circulating myostatin [41].
Another inhibitor of myostatin is growth and differentiation factor-associated serum protein 1 (GASP-1). Unlike members of the follistatin family, GASP-1 does not have affinity to activin [42]. The structure of GASP-1 is different from previously described inhibitors: With the exception of one follistatin domain, it has additional domains typical for protease inhibitors. Interestingly, GASP-1 can also bind to the myostatin propeptide and possibly regulate the activation of myostatin through proteolytic cleavage [42]. Moreover, myostatin can be inactivated upon covalent binding to latent TGF-β binding protein 3 (LTBP3). This inactive complex of myostatin/LTBP3 is used for myostatin storage in the extracellular matrix [50]. Taken together, myostatin is regulated by at least four different inhibitors via binding of the active or latent form. The mechanisms of myostatin inhibition still remain elusive, and the crosstalk between the different intrinsic inhibitors is still unclear. The variety of inhibitors emphasizes the importance of a strict myostatin regulation in order to avoid muscle damage and wasting. The schematic mechanism of myostatin extracellular signalling is presented in Fig. 1.

5 Intracellular response

Skeletal muscle mass is maintained as a consequence of two main molecular mechanisms: protein synthesis and protein degradation. Despite the profound knowledge of the role of myostatin in the regulation of muscle growth, many details of the molecular mechanisms of its action are poorly understood. It has been suggested that binding of myostatin to the ActRIIB results in the phosphorylation of two serine residues of Smad2 or Smad3 at COOH domains. This leads to the assembly of Smad2/3 with Smad4 to the heterodimer that is able to translocate to the nucleus and activate transcription of target genes [5153]. One of the known downstream targets of Smad signalling is MyoD, a transcriptional factor that is involved in skeletal muscle development and takes part in the repair of damaged skeletal muscle [5457]. Downregulation of myoD expression was shown in vitro and in vivo during cachexia, possibly via TNFα through the induction of the NF-κB pathway. Interestingly, myostatin downregulates myoD in an NF-κB-independent way [58, 59]. Myostatin also inhibits Pax3 expression, which is possibly an upstream target of MyoD [58, 60, 61]. Moreover, Smad signalling targets other genes such as myf5 and myogenin, known to be important for myogenesis [62].
Interestingly, other Smad proteins (Smad6 and Smad7) work as agonists. They compete for the binding to the activin type I receptor and thereby inhibit signalling of TGF-β family members [63, 64]. Myostatin was also shown to activate Smad7 transcription. Smad7, in turn, is able to inhibit the association of Smad2/3 with Smad4 [65, 66]. Therefore, the Smad pathway is regulated by feedback control [67].
Myostatin signalling via Smad has been intensively investigated, but it is not the only feasible signalling pathway. TGF-β family members were shown to activate mitogen-activated protein kinases (MAPKs), particularly p38 and extracellular signal-regulated kinase 1/2 (ERK1/2) [6871]. p38 MAPK is responsible for the cell response to stress factors and was shown to be activated by myostatin via TGF-β activated kinase 1 (TAK-1)/MAPK kinase (MAPKK) cascade. Its signalling results in the downregulation of myogenesis-related genes, but it does not target Smads [72]. The role of ERK1/2 in the regulation of muscle mass is controversial. On the one hand, there are studies confirming that ERK1/2 takes part in the process of satellite cell proliferation that is necessary for the maintenance of skeletal muscle weight [73] and induces protein synthesis under physiological conditions [74, 75]. On the other hand, there are experiments showing an opposite effect of ERK1/2 activation [7679]. Thus, the data suggest that increased activity of ERK1/2 leads to the differentiation inhibition in several cell types [76, 77]. At the same time, myostatin significantly activated ERK1/2 in C2C12 cells. Similar effects were observed in mice during systematic administration of myostatin [78]. Taken together, it seems likely that myostatin mediates its signal at least partially through ERK1/2 activation. Therefore, different responses through ERK1/2 could be caused by different levels of myostatin corresponding to normal and pathological conditions. MAPK cascade normally involves the activation of Ras/Raf/MEK1. To check whether myostatin uses the same pathways to activate ERK1/2, some experiments were done. Using an inhibitor of MEK1 in C2C12 cells, Yang et al. showed that this kinase is involved in the myostatin-induced activation of ERK1/2 [78]. Moreover, such inhibition of MEK1 leads to the rescue of cell differentiation, which means that MEK-1/ERK1/2 play a role in differentiation suppression by myostatin. The presence of dominant negative form of Ras was shown to positively influence MEK1/ERK1/2 through the downstream activation of Raf. Therefore, myostatin activates ERK1/2 via Ras/Raf/MEK1 pathway [78].

6 Cross talk of myostatin and IGF-1

One of the main positive regulators of muscle growth is insulin-like growth factor 1 (IGF-1). Under normal conditions, IGF-1 signalling seems to be dominant and blocks the myostatin pathway [80]. However, an inhibition of IGF-1 was observed when myostatin is overexpressed [81, 82]. IGF-1 can prevent TGF-β family-mediated apoptosis [83], and it was shown that in the absence of IGF-1, the level of apoptosis in C2C12 cells treated with myostatin increased. Yang et al. speculate it may in part explain the ability of myostatin in regulating cell cycle but not apoptosis in normal conditions [84].
The mechanism by which IGF-1 regulates myostatin signalling includes the inhibition of transcription factors responsible for the induction of atrogenes via phosphorylation with phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Akt plays a significant role in different metabolic processes in the cell, particularly in the hypertrophic response to insulin and IGF-1 [85, 86]. Akt is the crossing point between IGF-1/myostatin pathways. The results of several studies suggest that the genetic loss of myostatin leads to the increase in Akt activity in skeletal muscle in vivo and in vitro [82, 87]. In contrast, a decreased level of phosphorylated Akt is associated with incubation of myotubes with myostatin [58]. It is likely that in the conditions of muscle wasting, myostatin can switch Akt/mammalian target of rapamycin (mTOR) pathway, responsible for protein synthesis, to inhibition of protein synthesis involving FoxO, GSK-3β or other unknown patterns leading to the loss of muscle mass. These pathways will be discussed below in more detail.
Akt/FoxO
The class of Forkhead box O (FoxO) transcription factors is involved in the regulation of energy metabolism. FoxOs take part in the formation of skeletal muscle and adipose tissue as major organs for energy distribution [88, 89]. During myoblast proliferation, FoxO1 and FoxO3 are localized in the cytoplasm in the latent, phosphorylated form. Upon initiation of differentiation, FoxO translocates to the nucleus where it binds to DNA and regulates transcription. Akt regulates the activity of FoxO1 and FoxO3 by phosphorylating them and thereby retaining them in the cytoplasm [58]. In myotube culture, the level of FoxO1 in the nucleus was shown to increase after treatment with myostatin [58, 90], together with FoxO3 resulting in the induction of the muscle-specific E3-ubiquitin ligases atrogin-1 (MAFbx) and MuRF-1 [91, 92]. Expression of constitutively active FoxO1 in transgenic mice led to decrease in myoblast differentiation and a reduction in muscle weight [93]. In muscle, FoxOs are known to interact with Smad3 and Smad4 inducing the protein degradation. Recently, it was shown that FoxO1 and Smad synergistically increase the expression of myostatin mRNA and its promoter activity in C2C12 myotubes but via different pathways [94]. Taken together, myostatin-mediated signalling activates FoxO, and it leads to the upregulation of proteasome ubiquitin ligases MuRF-1 and atrogin-1, which participate in protein degradation.
Akt/GSK-3β
Cycline D1 is an important component for G1 phase of cell cycle which is also regulated via PI3K/Akt pathway. In C2C12 myoblasts treated with myostatin during the phase of active cell growth, a decrease in cycline D1 levels was shown, suggesting that myostatin targets cycline D1 for proliferation inhibition. In favour of this hypothesis, overexpression of cycline D1 was shown to rescue cell cycle. The activation of GSK-3β via dephosphorylation at Ser 9 increases proteolysis of cyclin D1 [95] in the response to myostatin-mediated dephosphorylation of Akt at Ser 473. Usage of IGF-1 or active Akt treatment blocks myostatin induced arrest of cell proliferation [84]. Thus, one more pathway activated by myostatin suggests that inhibition of Akt plays a dramatic role in the providing of myostatin signalling.
Akt/mTOR/p70s6K
This pathway is important for the differentiation of myoblasts and hypertrophy of myotubes. mTOR is a kinase downstream of Akt, which phosphorylates 4E-BP1 and p70s6k thereby inducing initiation of protein synthesis [85, 96]. In Mstn knockout mice, an increase in Akt/mTOR/p70s6K signalling was observed [87]. There are two mTOR complexes called TORC1 and TORC2, containing the proteins RAPTOR and RICTOR, respectively [97]. Activation of TORC1 leads to protein synthesis, while TORC2 plays a significant role in the feedback Akt phosphorylation leading to the blockade of FoxO signalling [98]. Inhibition of RAPTOR was shown to play an important role in myostatin signalling, whereas RICTOR is necessary for the cell differentiation itself. Inhibition of RAPTOR amplifies myostatin actions such as Smad phosphorylation [80].
Additional data confirming the connection between myostatin and Akt pathways were received in experiments with hypoxic muscles. The expression of myostatin was shown to be upregulated under these conditions [99]. At the same time, the Akt/mTOR pathway was downregulated in the rats with chronic hypoxia [100].
In conclusion, signal transduction of myostatin is a complex process involving activation and inhibition of several cellular signalling pathways. These pathways result in the downregulation of the expression of myogenic factors, decrease in protein synthesis and activation of proteasome–ubiquitin ligases. The schematic picture of myostatin signalling is presented in Fig. 2. There are a lot of questions still connected with the order of activation and precise importance of each pathway and its contribution in the development of muscle wasting.

7 Treatment

Myostatin has become a main target for the development of drugs for cachexia and muscle wasting diseases. Despite the experiments using antimyostatin substances that were conducted, there are no drug discoveries against muscle wasting in cachexia so far. The information about compounds that were involved in clinical trials is scarce, but none of the known substances had a positive effect. MYO-029 is recombinant human myostatin-specific antibody that did not show a significant improvement in muscle strength and function in adult muscular dystrophies [44]. The other compound developed by Amgen, AMG 745, has an unknown composition, but its testing was stopped after phase I clinical trials [102]. The problem of using antimyostatin compounds is likely due to the ability of the ActRIIB to bind other ligands from the TGF-β family resulting in a redundancy of myostatin.
The impact of sActRIIB use in the regulation of muscle mass is intensively studied at the moment. Recently, very positive results were published by Zhou et al. [46] in C-26 tumour-bearing mice, where the use of sActRIIB resulted in the prolongation of life and reversal of muscle wasting during cachexia. In this study, a positive effect on cardiac muscle was also seen. The results of this study provide a major step forward in establishing novel cachexia therapies.
The significance of these results for the treatment of cardiac cachexia was shown by Heineke et al. [27], who observed that myostatin produced from cardiomyocytes is released into the circulation in heart failure and that myostatin subsequently activated muscle wasting. The deletion of Mstn from cardiomyocytes and inhibition of myostatin protein using antibodies in mice prevented muscle wasting in heart failure [27].

8 Conclusion

Myostatin is a negative regulator of myoblast proliferation and differentiation. Normally it functions to regulate hypertrophy of muscles, but a role in the induction of muscle loss was observed in muscle wasting diseases and cachexia associated with severe illnesses. The mechanism of myostatin signalling is complex and involves the activation of several downstream pathways. Additional studies are required to elucidate the cross talk between the signalling cascades and their regulation. The experiments with myostatin propeptide and antimyostatin antibodies showed a positive effect on regulation of muscle mass in different models of wasting but lacked efficacy in phase I clinical trials. The most promising target in terms of cachexia treatment seems to be the activin type II receptor. The blockade of this receptor led to a significant increase in muscle mass and even its restoration in cancer cachexia. Therefore, targeting myostatin and its receptor represent a promising direction in the development of effective treatments for cachexia and muscle wasting diseases.

Acknowledgements

The authors of this manuscript certify that they comply with the ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle [103].

Conflict of interest

The authors declare no conflict of interest.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat von Haehling S, Morley JE, Anker SD. An overview of sarcopenia: facts and numbers on prevalence and clinical impact. J Cachexia Sarcopenia Muscle. 2010;1:129–33. von Haehling S, Morley JE, Anker SD. An overview of sarcopenia: facts and numbers on prevalence and clinical impact. J Cachexia Sarcopenia Muscle. 2010;1:129–33.
2.
Zurück zum Zitat Evans WJ, Morley JE, Argiles J, Bales C, Baracos V, Guttridge D, et al. Cachexia: a new definition. Clin Nutr. 2008;27:793–9.PubMed Evans WJ, Morley JE, Argiles J, Bales C, Baracos V, Guttridge D, et al. Cachexia: a new definition. Clin Nutr. 2008;27:793–9.PubMed
3.
Zurück zum Zitat Moldawer LL, Copeland 3rd EM. Proinflammatory cytokines, nutritional support, and the cachexia syndrome: interactions and therapeutic options. Cancer. 1997;79:1828–39.PubMed Moldawer LL, Copeland 3rd EM. Proinflammatory cytokines, nutritional support, and the cachexia syndrome: interactions and therapeutic options. Cancer. 1997;79:1828–39.PubMed
4.
Zurück zum Zitat Akamizu T, Kangawa K. Ghrelin for cachexia. J Cachexia Sarcopenia Muscle. 2010;1:169–76. Akamizu T, Kangawa K. Ghrelin for cachexia. J Cachexia Sarcopenia Muscle. 2010;1:169–76.
5.
Zurück zum Zitat Muller TD, Perez-Tilve D, Tong J, Pfluger PT, Tschop MH. Ghrelin and its potential in the treatment of eating/wasting disorders and cachexia. J Cachexia Sarcopenia Muscle. 2010;1:159–67. Muller TD, Perez-Tilve D, Tong J, Pfluger PT, Tschop MH. Ghrelin and its potential in the treatment of eating/wasting disorders and cachexia. J Cachexia Sarcopenia Muscle. 2010;1:159–67.
6.
Zurück zum Zitat von Haehling S, Lainscak M, Springer J, Anker SD. Cardiac cachexia: a systematic overview. Pharmacol Ther. 2009;121:227–52. von Haehling S, Lainscak M, Springer J, Anker SD. Cardiac cachexia: a systematic overview. Pharmacol Ther. 2009;121:227–52.
7.
Zurück zum Zitat McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387:83–90.PubMed McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature. 1997;387:83–90.PubMed
8.
Zurück zum Zitat Yarasheski KE, Bhasin S, Sinha-Hikim I, Pak-Loduca J, Gonzalez-Cadavid NF. Serum myostatin-immunoreactive protein is increased in 60–92 year old women and men with muscle wasting. J Nutr Health Aging. 2002;6:343–8.PubMed Yarasheski KE, Bhasin S, Sinha-Hikim I, Pak-Loduca J, Gonzalez-Cadavid NF. Serum myostatin-immunoreactive protein is increased in 60–92 year old women and men with muscle wasting. J Nutr Health Aging. 2002;6:343–8.PubMed
9.
Zurück zum Zitat Lenk K, Schur R, Linke A, Erbs S, Matsumoto Y, Adams V, et al. Impact of exercise training on myostatin expression in the myocardium and skeletal muscle in a chronic heart failure model. Eur J Heart Fail. 2009;11:342–8.PubMed Lenk K, Schur R, Linke A, Erbs S, Matsumoto Y, Adams V, et al. Impact of exercise training on myostatin expression in the myocardium and skeletal muscle in a chronic heart failure model. Eur J Heart Fail. 2009;11:342–8.PubMed
10.
Zurück zum Zitat Gonzalez-Cadavid NF, Taylor WE, Yarasheski K, Sinha-Hikim I, Ma K, Ezzat S, et al. Organization of the human myostatin gene and expression in healthy men and HIV-infected men with muscle wasting. Proc Natl Acad Sci USA. 1998;95:14938–43.PubMed Gonzalez-Cadavid NF, Taylor WE, Yarasheski K, Sinha-Hikim I, Ma K, Ezzat S, et al. Organization of the human myostatin gene and expression in healthy men and HIV-infected men with muscle wasting. Proc Natl Acad Sci USA. 1998;95:14938–43.PubMed
11.
Zurück zum Zitat Zimmers TA, Davies MV, Koniaris LG, Haynes P, Esquela AF, Tomkinson KN, et al. Induction of cachexia in mice by systemically administered myostatin. Science. 2002;296:1486–8.PubMed Zimmers TA, Davies MV, Koniaris LG, Haynes P, Esquela AF, Tomkinson KN, et al. Induction of cachexia in mice by systemically administered myostatin. Science. 2002;296:1486–8.PubMed
12.
Zurück zum Zitat Costelli P, Muscaritoli M, Bonetto A, Penna F, Reffo P, Bossola M, et al. Muscle myostatin signalling is enhanced in experimental cancer cachexia. Eur J Clin Invest. 2008;38:531–8.PubMed Costelli P, Muscaritoli M, Bonetto A, Penna F, Reffo P, Bossola M, et al. Muscle myostatin signalling is enhanced in experimental cancer cachexia. Eur J Clin Invest. 2008;38:531–8.PubMed
13.
Zurück zum Zitat Wolfman NM, McPherron AC, Pappano WN, Davies MV, Song K, Tomkinson KN, et al. Activation of latent myostatin by the BMP-1/tolloid family of metalloproteinases. Proc Natl Acad Sci USA. 2003;100:15842–6.PubMed Wolfman NM, McPherron AC, Pappano WN, Davies MV, Song K, Tomkinson KN, et al. Activation of latent myostatin by the BMP-1/tolloid family of metalloproteinases. Proc Natl Acad Sci USA. 2003;100:15842–6.PubMed
14.
Zurück zum Zitat McPherron AC, Lee SJ. Suppression of body fat accumulation in myostatin-deficient mice. J Clin Invest. 2002;109:595–601.PubMed McPherron AC, Lee SJ. Suppression of body fat accumulation in myostatin-deficient mice. J Clin Invest. 2002;109:595–601.PubMed
15.
Zurück zum Zitat Hamrick MW, Pennington C, Webb CN, Isales CM. Resistance to body fat gain in ‘double-muscled’ mice fed a high-fat diet. Int J Obes (Lond). 2006;30:868–70. Hamrick MW, Pennington C, Webb CN, Isales CM. Resistance to body fat gain in ‘double-muscled’ mice fed a high-fat diet. Int J Obes (Lond). 2006;30:868–70.
16.
Zurück zum Zitat Mosher DS, Quignon P, Bustamante CD, Sutter NB, Mellersh CS, Parker HG, et al. A mutation in the myostatin gene increases muscle mass and enhances racing performance in heterozygote dogs. PLoS Genet. 2007;3:e79.PubMed Mosher DS, Quignon P, Bustamante CD, Sutter NB, Mellersh CS, Parker HG, et al. A mutation in the myostatin gene increases muscle mass and enhances racing performance in heterozygote dogs. PLoS Genet. 2007;3:e79.PubMed
17.
Zurück zum Zitat Grobet L, Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, et al. A deletion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nat Genet. 1997;17:71–4.PubMed Grobet L, Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, et al. A deletion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nat Genet. 1997;17:71–4.PubMed
18.
Zurück zum Zitat McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proc Natl Acad Sci USA. 1997;94:12457–61.PubMed McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proc Natl Acad Sci USA. 1997;94:12457–61.PubMed
19.
Zurück zum Zitat Clop A, Marcq F, Takeda H, Pirottin D, Tordoir X, Bibe B, et al. A mutation creating a potential illegitimate microRNA target site in the myostatin gene affects muscularity in sheep. Nat Genet. 2006;38:813–8.PubMed Clop A, Marcq F, Takeda H, Pirottin D, Tordoir X, Bibe B, et al. A mutation creating a potential illegitimate microRNA target site in the myostatin gene affects muscularity in sheep. Nat Genet. 2006;38:813–8.PubMed
20.
Zurück zum Zitat Lee SJ. Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol. 2004;20:61–86.PubMed Lee SJ. Regulation of muscle mass by myostatin. Annu Rev Cell Dev Biol. 2004;20:61–86.PubMed
21.
Zurück zum Zitat Morissette MR, Cook SA, Foo S, McKoy G, Ashida N, Novikov M, et al. Myostatin regulates cardiomyocyte growth through modulation of Akt signaling. Circ Res. 2006;99:15–24.PubMed Morissette MR, Cook SA, Foo S, McKoy G, Ashida N, Novikov M, et al. Myostatin regulates cardiomyocyte growth through modulation of Akt signaling. Circ Res. 2006;99:15–24.PubMed
22.
Zurück zum Zitat Sharma M, Kambadur R, Matthews KG, Somers WG, Devlin GP, Conaglen JV, et al. Myostatin, a transforming growth factor-beta superfamily member, is expressed in heart muscle and is upregulated in cardiomyocytes after infarct. J Cell Physiol. 1999;180:1–9.PubMed Sharma M, Kambadur R, Matthews KG, Somers WG, Devlin GP, Conaglen JV, et al. Myostatin, a transforming growth factor-beta superfamily member, is expressed in heart muscle and is upregulated in cardiomyocytes after infarct. J Cell Physiol. 1999;180:1–9.PubMed
23.
Zurück zum Zitat Shyu KG, Lu MJ, Wang BW, Sun HY, Chang H. Myostatin expression in ventricular myocardium in a rat model of volume-overload heart failure. Eur J Clin Invest. 2006;36:713–9.PubMed Shyu KG, Lu MJ, Wang BW, Sun HY, Chang H. Myostatin expression in ventricular myocardium in a rat model of volume-overload heart failure. Eur J Clin Invest. 2006;36:713–9.PubMed
24.
Zurück zum Zitat Allen DL, Cleary AS, Speaker KJ, Lindsay SF, Uyenishi J, Reed JM, et al. Myostatin, activin receptor IIb, and follistatin-like-3 gene expression are altered in adipose tissue and skeletal muscle of obese mice. Am J Physiol Endocrinol Metab. 2008;294:E918–27.PubMed Allen DL, Cleary AS, Speaker KJ, Lindsay SF, Uyenishi J, Reed JM, et al. Myostatin, activin receptor IIb, and follistatin-like-3 gene expression are altered in adipose tissue and skeletal muscle of obese mice. Am J Physiol Endocrinol Metab. 2008;294:E918–27.PubMed
25.
Zurück zum Zitat Ji S, Losinski RL, Cornelius SG, Frank GR, Willis GM, Gerrard DE, et al. Myostatin expression in porcine tissues: tissue specificity and developmental and postnatal regulation. Am J Physiol. 1998;275:R1265–73.PubMed Ji S, Losinski RL, Cornelius SG, Frank GR, Willis GM, Gerrard DE, et al. Myostatin expression in porcine tissues: tissue specificity and developmental and postnatal regulation. Am J Physiol. 1998;275:R1265–73.PubMed
26.
Zurück zum Zitat George I, Bish LT, Kamalakkannan G, Petrilli CM, Oz MC, Naka Y, et al. Myostatin activation in patients with advanced heart failure and after mechanical unloading. Eur J Heart Fail. 2010;12:444–53.PubMed George I, Bish LT, Kamalakkannan G, Petrilli CM, Oz MC, Naka Y, et al. Myostatin activation in patients with advanced heart failure and after mechanical unloading. Eur J Heart Fail. 2010;12:444–53.PubMed
27.
Zurück zum Zitat Heineke J, Auger-Messier M, Xu J, Sargent M, York A, Welle S, et al. Genetic deletion of myostatin from the heart prevents skeletal muscle atrophy in heart failure. Circulation. 2010;121:419–25.PubMed Heineke J, Auger-Messier M, Xu J, Sargent M, York A, Welle S, et al. Genetic deletion of myostatin from the heart prevents skeletal muscle atrophy in heart failure. Circulation. 2010;121:419–25.PubMed
28.
Zurück zum Zitat Rebbapragada A, Benchabane H, Wrana JL, Celeste AJ, Attisano L. Myostatin signals through a transforming growth factor beta-like signaling pathway to block adipogenesis. Mol Cell Biol. 2003;23:7230–42.PubMed Rebbapragada A, Benchabane H, Wrana JL, Celeste AJ, Attisano L. Myostatin signals through a transforming growth factor beta-like signaling pathway to block adipogenesis. Mol Cell Biol. 2003;23:7230–42.PubMed
29.
Zurück zum Zitat Kim HS, Liang L, Dean RG, Hausman DB, Hartzell DL, Baile CA. Inhibition of preadipocyte differentiation by myostatin treatment in 3T3-L1 cultures. Biochem Biophys Res Commun. 2001;281:902–6.PubMed Kim HS, Liang L, Dean RG, Hausman DB, Hartzell DL, Baile CA. Inhibition of preadipocyte differentiation by myostatin treatment in 3T3-L1 cultures. Biochem Biophys Res Commun. 2001;281:902–6.PubMed
30.
Zurück zum Zitat Artaza JN, Bhasin S, Magee TR, Reisz-Porszasz S, Shen R, Groome NP, et al. Myostatin inhibits myogenesis and promotes adipogenesis in C3H 10T(1/2) mesenchymal multipotent cells. Endocrinology. 2005;146:3547–57.PubMed Artaza JN, Bhasin S, Magee TR, Reisz-Porszasz S, Shen R, Groome NP, et al. Myostatin inhibits myogenesis and promotes adipogenesis in C3H 10T(1/2) mesenchymal multipotent cells. Endocrinology. 2005;146:3547–57.PubMed
31.
Zurück zum Zitat Feldman BJ, Streeper RS, Farese Jr RV, Yamamoto KR. Myostatin modulates adipogenesis to generate adipocytes with favorable metabolic effects. Proc Natl Acad Sci USA. 2006;103:15675–80.PubMed Feldman BJ, Streeper RS, Farese Jr RV, Yamamoto KR. Myostatin modulates adipogenesis to generate adipocytes with favorable metabolic effects. Proc Natl Acad Sci USA. 2006;103:15675–80.PubMed
32.
Zurück zum Zitat Lin J, Arnold HB, Della-Fera MA, Azain MJ, Hartzell DL, Baile CA. Myostatin knockout in mice increases myogenesis and decreases adipogenesis. Biochem Biophys Res Commun. 2002;291:701–6.PubMed Lin J, Arnold HB, Della-Fera MA, Azain MJ, Hartzell DL, Baile CA. Myostatin knockout in mice increases myogenesis and decreases adipogenesis. Biochem Biophys Res Commun. 2002;291:701–6.PubMed
33.
Zurück zum Zitat Lai KM, Gonzalez M, Poueymirou WT, Kline WO, Na E, Zlotchenko E, et al. Conditional activation of Akt in adult skeletal muscle induces rapid hypertrophy. Mol Cell Biol. 2004;24:9295–304.PubMed Lai KM, Gonzalez M, Poueymirou WT, Kline WO, Na E, Zlotchenko E, et al. Conditional activation of Akt in adult skeletal muscle induces rapid hypertrophy. Mol Cell Biol. 2004;24:9295–304.PubMed
34.
Zurück zum Zitat Izumiya Y, Hopkins T, Morris C, Sato K, Zeng L, Viereck J, et al. Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell Metab. 2008;7:159–72.PubMed Izumiya Y, Hopkins T, Morris C, Sato K, Zeng L, Viereck J, et al. Fast/Glycolytic muscle fiber growth reduces fat mass and improves metabolic parameters in obese mice. Cell Metab. 2008;7:159–72.PubMed
35.
Zurück zum Zitat Musaro A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, et al. Localized IGF-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet. 2001;27:195–200.PubMed Musaro A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, et al. Localized IGF-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet. 2001;27:195–200.PubMed
36.
Zurück zum Zitat Lee SJ, McPherron AC. Regulation of myostatin activity and muscle growth. Proc Natl Acad Sci USA. 2001;98:9306–11.PubMed Lee SJ, McPherron AC. Regulation of myostatin activity and muscle growth. Proc Natl Acad Sci USA. 2001;98:9306–11.PubMed
37.
Zurück zum Zitat Kung T, Szabo T, Springer J, Doehner W, Anker SD, von Haehling S. Cachexia in heart disease: highlights from the ESC 2010. J Cachexia Sarcopenia Muscle. 2011;2:63–9. Kung T, Szabo T, Springer J, Doehner W, Anker SD, von Haehling S. Cachexia in heart disease: highlights from the ESC 2010. J Cachexia Sarcopenia Muscle. 2011;2:63–9.
38.
Zurück zum Zitat Sako D, Grinberg AV, Liu J, Davies MV, Castonguay R, Maniatis S, et al. Characterization of the ligand binding functionality of the extracellular domain of activin receptor type IIb. J Biol Chem. 2010;285:21037–48.PubMed Sako D, Grinberg AV, Liu J, Davies MV, Castonguay R, Maniatis S, et al. Characterization of the ligand binding functionality of the extracellular domain of activin receptor type IIb. J Biol Chem. 2010;285:21037–48.PubMed
39.
Zurück zum Zitat Wrana JL, Attisano L, Wieser R, Ventura F, Massague J. Mechanism of activation of the TGF-beta receptor. Nature. 1994;370:341–7.PubMed Wrana JL, Attisano L, Wieser R, Ventura F, Massague J. Mechanism of activation of the TGF-beta receptor. Nature. 1994;370:341–7.PubMed
40.
Zurück zum Zitat Lee SJ, Reed LA, Davies MV, Girgenrath S, Goad ME, Tomkinson KN, et al. Regulation of muscle growth by multiple ligands signaling through activin type II receptors. Proc Natl Acad Sci USA. 2005;102:18117–22.PubMed Lee SJ, Reed LA, Davies MV, Girgenrath S, Goad ME, Tomkinson KN, et al. Regulation of muscle growth by multiple ligands signaling through activin type II receptors. Proc Natl Acad Sci USA. 2005;102:18117–22.PubMed
41.
Zurück zum Zitat Hill JJ, Davies MV, Pearson AA, Wang JH, Hewick RM, Wolfman NM, et al. The myostatin propeptide and the follistatin-related gene are inhibitory binding proteins of myostatin in normal serum. J Biol Chem. 2002;277:40735–41.PubMed Hill JJ, Davies MV, Pearson AA, Wang JH, Hewick RM, Wolfman NM, et al. The myostatin propeptide and the follistatin-related gene are inhibitory binding proteins of myostatin in normal serum. J Biol Chem. 2002;277:40735–41.PubMed
42.
Zurück zum Zitat Hill JJ, Qiu Y, Hewick RM, Wolfman NM. Regulation of myostatin in vivo by growth and differentiation factor-associated serum protein-1: a novel protein with protease inhibitor and follistatin domains. Mol Endocrinol. 2003;17:1144–54.PubMed Hill JJ, Qiu Y, Hewick RM, Wolfman NM. Regulation of myostatin in vivo by growth and differentiation factor-associated serum protein-1: a novel protein with protease inhibitor and follistatin domains. Mol Endocrinol. 2003;17:1144–54.PubMed
43.
Zurück zum Zitat Souza TA, Chen X, Guo Y, Sava P, Zhang J, Hill JJ, et al. Proteomic identification and functional validation of activins and bone morphogenetic protein 11 as candidate novel muscle mass regulators. Mol Endocrinol. 2008;22:2689–702.PubMed Souza TA, Chen X, Guo Y, Sava P, Zhang J, Hill JJ, et al. Proteomic identification and functional validation of activins and bone morphogenetic protein 11 as candidate novel muscle mass regulators. Mol Endocrinol. 2008;22:2689–702.PubMed
44.
Zurück zum Zitat Wagner KR, Fleckenstein JL, Amato AA, Barohn RJ, Bushby K, Escolar DM, et al. A phase I/II trial of MYO-029 in adult subjects with muscular dystrophy. Ann Neurol. 2008;63:561–71.PubMed Wagner KR, Fleckenstein JL, Amato AA, Barohn RJ, Bushby K, Escolar DM, et al. A phase I/II trial of MYO-029 in adult subjects with muscular dystrophy. Ann Neurol. 2008;63:561–71.PubMed
45.
Zurück zum Zitat Benny Klimek ME, Aydogdu T, Link MJ, Pons M, Koniaris LG, Zimmers TA. Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia. Biochem Biophys Res Commun. 2010;391:1548–54.PubMed Benny Klimek ME, Aydogdu T, Link MJ, Pons M, Koniaris LG, Zimmers TA. Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia. Biochem Biophys Res Commun. 2010;391:1548–54.PubMed
46.
Zurück zum Zitat Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell. 2010;142:531–43.PubMed Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell. 2010;142:531–43.PubMed
47.
Zurück zum Zitat Thies RS, Chen T, Davies MV, Tomkinson KN, Pearson AA, Shakey QA, et al. GDF-8 propeptide binds to GDF-8 and antagonizes biological activity by inhibiting GDF-8 receptor binding. Growth Factors. 2001;18:251–9.PubMed Thies RS, Chen T, Davies MV, Tomkinson KN, Pearson AA, Shakey QA, et al. GDF-8 propeptide binds to GDF-8 and antagonizes biological activity by inhibiting GDF-8 receptor binding. Growth Factors. 2001;18:251–9.PubMed
48.
Zurück zum Zitat Yang J, Ratovitski T, Brady JP, Solomon MB, Wells KD, Wall RJ. Expression of myostatin pro domain results in muscular transgenic mice. Mol Reprod Dev. 2001;60:351–61.PubMed Yang J, Ratovitski T, Brady JP, Solomon MB, Wells KD, Wall RJ. Expression of myostatin pro domain results in muscular transgenic mice. Mol Reprod Dev. 2001;60:351–61.PubMed
49.
Zurück zum Zitat Matzuk MM, Lu N, Vogel H, Sellheyer K, Roop DR, Bradley A. Multiple defects and perinatal death in mice deficient in follistatin. Nature. 1995;374:360–3.PubMed Matzuk MM, Lu N, Vogel H, Sellheyer K, Roop DR, Bradley A. Multiple defects and perinatal death in mice deficient in follistatin. Nature. 1995;374:360–3.PubMed
50.
Zurück zum Zitat Anderson SB, Goldberg AL, Whitman M. Identification of a novel pool of extracellular pro-myostatin in skeletal muscle. J Biol Chem. 2008;283:7027–35.PubMed Anderson SB, Goldberg AL, Whitman M. Identification of a novel pool of extracellular pro-myostatin in skeletal muscle. J Biol Chem. 2008;283:7027–35.PubMed
51.
Zurück zum Zitat Joulia-Ekaza D, Cabello G. The myostatin gene: physiology and pharmacological relevance. Curr Opin Pharmacol. 2007;7:310–5.PubMed Joulia-Ekaza D, Cabello G. The myostatin gene: physiology and pharmacological relevance. Curr Opin Pharmacol. 2007;7:310–5.PubMed
52.
Zurück zum Zitat Kollias HD, McDermott JC. Transforming growth factor-beta and myostatin signaling in skeletal muscle. J Appl Physiol. 2008;104:579–87.PubMed Kollias HD, McDermott JC. Transforming growth factor-beta and myostatin signaling in skeletal muscle. J Appl Physiol. 2008;104:579–87.PubMed
53.
Zurück zum Zitat Patel K, Amthor H. The function of myostatin and strategies of myostatin blockade-new hope for therapies aimed at promoting growth of skeletal muscle. Neuromuscul Disord. 2005;15:117–26.PubMed Patel K, Amthor H. The function of myostatin and strategies of myostatin blockade-new hope for therapies aimed at promoting growth of skeletal muscle. Neuromuscul Disord. 2005;15:117–26.PubMed
54.
Zurück zum Zitat Megeney LA, Kablar B, Garrett K, Anderson JE, Rudnicki MA. MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev. 1996;10:1173–83.PubMed Megeney LA, Kablar B, Garrett K, Anderson JE, Rudnicki MA. MyoD is required for myogenic stem cell function in adult skeletal muscle. Genes Dev. 1996;10:1173–83.PubMed
55.
Zurück zum Zitat Cornelison DD, Olwin BB, Rudnicki MA, Wold BJ. MyoD(−/−) satellite cells in single-fiber culture are differentiation defective and MRF4 deficient. Dev Biol. 2000;224:122–37.PubMed Cornelison DD, Olwin BB, Rudnicki MA, Wold BJ. MyoD(−/−) satellite cells in single-fiber culture are differentiation defective and MRF4 deficient. Dev Biol. 2000;224:122–37.PubMed
56.
Zurück zum Zitat Guttridge DC, Mayo MW, Madrid LV, Wang CY, Baldwin Jr AS. NF-kappaB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science. 2000;289:2363–6.PubMed Guttridge DC, Mayo MW, Madrid LV, Wang CY, Baldwin Jr AS. NF-kappaB-induced loss of MyoD messenger RNA: possible role in muscle decay and cachexia. Science. 2000;289:2363–6.PubMed
57.
Zurück zum Zitat Montarras D, Lindon C, Pinset C, Domeyne P. Cultured myf5 null and myoD null muscle precursor cells display distinct growth defects. Biol Cell. 2000;92:565–72.PubMed Montarras D, Lindon C, Pinset C, Domeyne P. Cultured myf5 null and myoD null muscle precursor cells display distinct growth defects. Biol Cell. 2000;92:565–72.PubMed
58.
Zurück zum Zitat McFarlane C, Plummer E, Thomas M, Hennebry A, Ashby M, Ling N, et al. Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1-dependent mechanism. J Cell Physiol. 2006;209:501–14.PubMed McFarlane C, Plummer E, Thomas M, Hennebry A, Ashby M, Ling N, et al. Myostatin induces cachexia by activating the ubiquitin proteolytic system through an NF-kappaB-independent, FoxO1-dependent mechanism. J Cell Physiol. 2006;209:501–14.PubMed
59.
Zurück zum Zitat Bakkar N, Wackerhage H, Guttridge D. Myostatin and NF-κB regulate skeletal myogenesis through distinct signaling pathways. Signal Transduction. 2005;5:202–10. Bakkar N, Wackerhage H, Guttridge D. Myostatin and NF-κB regulate skeletal myogenesis through distinct signaling pathways. Signal Transduction. 2005;5:202–10.
60.
Zurück zum Zitat Tajbakhsh S, Rocancourt D, Cossu G, Buckingham M. Redefining the genetic hierarchies controlling skeletal myogenesis: Pax-3 and Myf-5 act upstream of MyoD. Cell. 1997;89:127–38.PubMed Tajbakhsh S, Rocancourt D, Cossu G, Buckingham M. Redefining the genetic hierarchies controlling skeletal myogenesis: Pax-3 and Myf-5 act upstream of MyoD. Cell. 1997;89:127–38.PubMed
61.
Zurück zum Zitat Maroto M, Reshef R, Munsterberg AE, Koester S, Goulding M, Lassar AB. Ectopic Pax-3 activates MyoD and Myf-5 expression in embryonic mesoderm and neural tissue. Cell. 1997;89:139–48.PubMed Maroto M, Reshef R, Munsterberg AE, Koester S, Goulding M, Lassar AB. Ectopic Pax-3 activates MyoD and Myf-5 expression in embryonic mesoderm and neural tissue. Cell. 1997;89:139–48.PubMed
62.
Zurück zum Zitat Langley B, Thomas M, Bishop A, Sharma M, Gilmour S, Kambadur R. Myostatin inhibits myoblast differentiation by down-regulating MyoD expression. J Biol Chem. 2002;277:49831–40.PubMed Langley B, Thomas M, Bishop A, Sharma M, Gilmour S, Kambadur R. Myostatin inhibits myoblast differentiation by down-regulating MyoD expression. J Biol Chem. 2002;277:49831–40.PubMed
63.
Zurück zum Zitat Chen Y, Lebrun JJ, Vale W. Regulation of transforming growth factor beta- and activin-induced transcription by mammalian Mad proteins. Proc Natl Acad Sci USA. 1996;93:12992–7.PubMed Chen Y, Lebrun JJ, Vale W. Regulation of transforming growth factor beta- and activin-induced transcription by mammalian Mad proteins. Proc Natl Acad Sci USA. 1996;93:12992–7.PubMed
64.
Zurück zum Zitat Zhao J, Shi W, Chen H, Warburton D. Smad7 and Smad6 differentially modulate transforming growth factor beta-induced inhibition of embryonic lung morphogenesis. J Biol Chem. 2000;275:23992–7.PubMed Zhao J, Shi W, Chen H, Warburton D. Smad7 and Smad6 differentially modulate transforming growth factor beta-induced inhibition of embryonic lung morphogenesis. J Biol Chem. 2000;275:23992–7.PubMed
65.
Zurück zum Zitat Nakao A, Imamura T, Souchelnytskyi S, Kawabata M, Ishisaki A, Oeda E, et al. TGF-beta receptor-mediated signalling through Smad2, Smad3 and Smad4. EMBO J. 1997;16:5353–62.PubMed Nakao A, Imamura T, Souchelnytskyi S, Kawabata M, Ishisaki A, Oeda E, et al. TGF-beta receptor-mediated signalling through Smad2, Smad3 and Smad4. EMBO J. 1997;16:5353–62.PubMed
66.
Zurück zum Zitat Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, et al. The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling. Cell. 1997;89:1165–73.PubMed Hayashi H, Abdollah S, Qiu Y, Cai J, Xu YY, Grinnell BW, et al. The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling. Cell. 1997;89:1165–73.PubMed
67.
Zurück zum Zitat Forbes D, Jackman M, Bishop A, Thomas M, Kambadur R, Sharma M. Myostatin auto-regulates its expression by feedback loop through Smad7 dependent mechanism. J Cell Physiol. 2006;206:264–72.PubMed Forbes D, Jackman M, Bishop A, Thomas M, Kambadur R, Sharma M. Myostatin auto-regulates its expression by feedback loop through Smad7 dependent mechanism. J Cell Physiol. 2006;206:264–72.PubMed
68.
Zurück zum Zitat Hartsough MT, Mulder KM. Transforming growth factor beta activation of p44mapk in proliferating cultures of epithelial cells. J Biol Chem. 1995;270:7117–24.PubMed Hartsough MT, Mulder KM. Transforming growth factor beta activation of p44mapk in proliferating cultures of epithelial cells. J Biol Chem. 1995;270:7117–24.PubMed
69.
Zurück zum Zitat Mulder KM. Role of Ras and Mapks in TGFbeta signaling. Cytokine Growth Factor Rev. 2000;11:23–35.PubMed Mulder KM. Role of Ras and Mapks in TGFbeta signaling. Cytokine Growth Factor Rev. 2000;11:23–35.PubMed
70.
Zurück zum Zitat Hanafusa H, Ninomiya-Tsuji J, Masuyama N, Nishita M, Fujisawa J, Shibuya H, et al. Involvement of the p38 mitogen-activated protein kinase pathway in transforming growth factor-beta-induced gene expression. J Biol Chem. 1999;274:27161–7.PubMed Hanafusa H, Ninomiya-Tsuji J, Masuyama N, Nishita M, Fujisawa J, Shibuya H, et al. Involvement of the p38 mitogen-activated protein kinase pathway in transforming growth factor-beta-induced gene expression. J Biol Chem. 1999;274:27161–7.PubMed
71.
Zurück zum Zitat Yu L, Hebert MC, Zhang YE. TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses. EMBO J. 2002;21:3749–59.PubMed Yu L, Hebert MC, Zhang YE. TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses. EMBO J. 2002;21:3749–59.PubMed
72.
Zurück zum Zitat Philip B, Lu Z, Gao Y. Regulation of GDF-8 signaling by the p38 MAPK. Cell Signal. 2005;17:365–75.PubMed Philip B, Lu Z, Gao Y. Regulation of GDF-8 signaling by the p38 MAPK. Cell Signal. 2005;17:365–75.PubMed
73.
Zurück zum Zitat Nicolas N, Marazzi G, Kelley K, Sassoon D. Embryonic deregulation of muscle stress signaling pathways leads to altered postnatal stem cell behavior and a failure in postnatal muscle growth. Dev Biol. 2005;281:171–83.PubMed Nicolas N, Marazzi G, Kelley K, Sassoon D. Embryonic deregulation of muscle stress signaling pathways leads to altered postnatal stem cell behavior and a failure in postnatal muscle growth. Dev Biol. 2005;281:171–83.PubMed
74.
Zurück zum Zitat Sale EM, Atkinson PP, Arnott CH, Chad JE, Sale GJ. Role of ERK1/ERK2 and p70S6K pathway in insulin signalling of protein synthesis. FEBS Lett. 1999;446:122–6.PubMed Sale EM, Atkinson PP, Arnott CH, Chad JE, Sale GJ. Role of ERK1/ERK2 and p70S6K pathway in insulin signalling of protein synthesis. FEBS Lett. 1999;446:122–6.PubMed
75.
Zurück zum Zitat Felton-Edkins ZA, Fairley JA, Graham EL, Johnston IM, White RJ, Scott PH. The mitogen-activated protein (MAP) kinase ERK induces tRNA synthesis by phosphorylating TFIIIB. EMBO J. 2003;22:2422–32.PubMed Felton-Edkins ZA, Fairley JA, Graham EL, Johnston IM, White RJ, Scott PH. The mitogen-activated protein (MAP) kinase ERK induces tRNA synthesis by phosphorylating TFIIIB. EMBO J. 2003;22:2422–32.PubMed
76.
Zurück zum Zitat Penn BH, Berkes CA, Bergstrom DA, Tapscott SJ. How to MEK muscle. Mol Cell. 2001;8:245–6.PubMed Penn BH, Berkes CA, Bergstrom DA, Tapscott SJ. How to MEK muscle. Mol Cell. 2001;8:245–6.PubMed
77.
Zurück zum Zitat Kang CD, Do IR, Kim KW, Ahn BK, Kim SH, Chung BS, et al. Role of Ras/ERK-dependent pathway in the erythroid differentiation of K562 cells. Exp Mol Med. 1999;31:76–82.PubMed Kang CD, Do IR, Kim KW, Ahn BK, Kim SH, Chung BS, et al. Role of Ras/ERK-dependent pathway in the erythroid differentiation of K562 cells. Exp Mol Med. 1999;31:76–82.PubMed
78.
Zurück zum Zitat Yang W, Chen Y, Zhang Y, Wang X, Yang N, Zhu D. Extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway is involved in myostatin-regulated differentiation repression. Cancer Res. 2006;66:1320–6.PubMed Yang W, Chen Y, Zhang Y, Wang X, Yang N, Zhu D. Extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway is involved in myostatin-regulated differentiation repression. Cancer Res. 2006;66:1320–6.PubMed
79.
Zurück zum Zitat Penna F, Costamagna D, Bonetto A, Minero VG, Baccino FM, Costelli P. Extracellular signal-regulated kinase (ERK) inhibition prevents muscle atrophy in experimental cancer cachexia. J Cachexia Sarcopenia Muscle. 2010;1:67. Abstract 054. Penna F, Costamagna D, Bonetto A, Minero VG, Baccino FM, Costelli P. Extracellular signal-regulated kinase (ERK) inhibition prevents muscle atrophy in experimental cancer cachexia. J Cachexia Sarcopenia Muscle. 2010;1:67. Abstract 054.
80.
Zurück zum Zitat Trendelenburg AU, Meyer A, Rohner D, Boyle J, Hatakeyama S, Glass DJ. Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Physiol Cell Physiol. 2009;296:C1258–70.PubMed Trendelenburg AU, Meyer A, Rohner D, Boyle J, Hatakeyama S, Glass DJ. Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Physiol Cell Physiol. 2009;296:C1258–70.PubMed
81.
Zurück zum Zitat Amirouche A, Durieux AC, Banzet S, Koulmann N, Bonnefoy R, Mouret C, et al. Down-regulation of Akt/mammalian target of rapamycin signaling pathway in response to myostatin overexpression in skeletal muscle. Endocrinology. 2009;150:286–94.PubMed Amirouche A, Durieux AC, Banzet S, Koulmann N, Bonnefoy R, Mouret C, et al. Down-regulation of Akt/mammalian target of rapamycin signaling pathway in response to myostatin overexpression in skeletal muscle. Endocrinology. 2009;150:286–94.PubMed
82.
Zurück zum Zitat Morissette MR, Cook SA, Buranasombati C, Rosenberg MA, Rosenzweig A. Myostatin inhibits IGF-I-induced myotube hypertrophy through Akt. Am J Physiol Cell Physiol. 2009;297:C1124–32.PubMed Morissette MR, Cook SA, Buranasombati C, Rosenberg MA, Rosenzweig A. Myostatin inhibits IGF-I-induced myotube hypertrophy through Akt. Am J Physiol Cell Physiol. 2009;297:C1124–32.PubMed
83.
Zurück zum Zitat Chen RH, Su YH, Chuang RL, Chang TY. Suppression of transforming growth factor-beta-induced apoptosis through a phosphatidylinositol 3-kinase/Akt-dependent pathway. Oncogene. 1998;17:1959–68.PubMed Chen RH, Su YH, Chuang RL, Chang TY. Suppression of transforming growth factor-beta-induced apoptosis through a phosphatidylinositol 3-kinase/Akt-dependent pathway. Oncogene. 1998;17:1959–68.PubMed
84.
Zurück zum Zitat Yang W, Zhang Y, Li Y, Wu Z, Zhu D. Myostatin induces cyclin D1 degradation to cause cell cycle arrest through a phosphatidylinositol 3-kinase/AKT/GSK-3 beta pathway and is antagonized by insulin-like growth factor 1. J Biol Chem. 2007;282:3799–808.PubMed Yang W, Zhang Y, Li Y, Wu Z, Zhu D. Myostatin induces cyclin D1 degradation to cause cell cycle arrest through a phosphatidylinositol 3-kinase/AKT/GSK-3 beta pathway and is antagonized by insulin-like growth factor 1. J Biol Chem. 2007;282:3799–808.PubMed
85.
Zurück zum Zitat Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 2001;3:1014–9.PubMed Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, et al. Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo. Nat Cell Biol. 2001;3:1014–9.PubMed
86.
Zurück zum Zitat Zdychova J, Komers R. Emerging role of Akt kinase/protein kinase B signaling in pathophysiology of diabetes and its complications. Physiol Res. 2005;54:1–16.PubMed Zdychova J, Komers R. Emerging role of Akt kinase/protein kinase B signaling in pathophysiology of diabetes and its complications. Physiol Res. 2005;54:1–16.PubMed
87.
Zurück zum Zitat Lipina C, Kendall H, McPherron AC, Taylor PM, Hundal HS. Mechanisms involved in the enhancement of mammalian target of rapamycin signalling and hypertrophy in skeletal muscle of myostatin-deficient mice. FEBS Lett. 2010;584:2403–8.PubMed Lipina C, Kendall H, McPherron AC, Taylor PM, Hundal HS. Mechanisms involved in the enhancement of mammalian target of rapamycin signalling and hypertrophy in skeletal muscle of myostatin-deficient mice. FEBS Lett. 2010;584:2403–8.PubMed
88.
Zurück zum Zitat Hribal ML, Nakae J, Kitamura T, Shutter JR, Accili D. Regulation of insulin-like growth factor-dependent myoblast differentiation by Foxo forkhead transcription factors. J Cell Biol. 2003;162:535–41.PubMed Hribal ML, Nakae J, Kitamura T, Shutter JR, Accili D. Regulation of insulin-like growth factor-dependent myoblast differentiation by Foxo forkhead transcription factors. J Cell Biol. 2003;162:535–41.PubMed
89.
Zurück zum Zitat Nakae J, Kitamura T, Kitamura Y, Biggs 3rd WH, Arden KC, Accili D. The forkhead transcription factor Foxo1 regulates adipocyte differentiation. Dev Cell. 2003;4:119–29.PubMed Nakae J, Kitamura T, Kitamura Y, Biggs 3rd WH, Arden KC, Accili D. The forkhead transcription factor Foxo1 regulates adipocyte differentiation. Dev Cell. 2003;4:119–29.PubMed
90.
Zurück zum Zitat Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell. 2004;117:399–412.PubMed Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell. 2004;117:399–412.PubMed
91.
Zurück zum Zitat Cohen S, Brault JJ, Gygi SP, Glass DJ, Valenzuela DM, Gartner C, et al. During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation. J Cell Biol. 2009;185:1083–95.PubMed Cohen S, Brault JJ, Gygi SP, Glass DJ, Valenzuela DM, Gartner C, et al. During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation. J Cell Biol. 2009;185:1083–95.PubMed
92.
Zurück zum Zitat Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab. 2007;6:472–83.PubMed Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab. 2007;6:472–83.PubMed
93.
Zurück zum Zitat Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Taniguchi T, et al. Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem. 2004;279:41114–23.PubMed Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Taniguchi T, et al. Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem. 2004;279:41114–23.PubMed
94.
Zurück zum Zitat Allen DL, Unterman TG. Regulation of myostatin expression and myoblast differentiation by FoxO and SMAD transcription factors. Am J Physiol Cell Physiol. 2007;292:C188–99.PubMed Allen DL, Unterman TG. Regulation of myostatin expression and myoblast differentiation by FoxO and SMAD transcription factors. Am J Physiol Cell Physiol. 2007;292:C188–99.PubMed
95.
Zurück zum Zitat Chen B, Pan H, Zhu L, Deng Y, Pollard JW. Progesterone inhibits the estrogen-induced phosphoinositide 3-kinase-->AKT-->GSK-3beta-->cyclin D1-->pRB pathway to block uterine epithelial cell proliferation. Mol Endocrinol. 2005;19:1978–90.PubMed Chen B, Pan H, Zhu L, Deng Y, Pollard JW. Progesterone inhibits the estrogen-induced phosphoinositide 3-kinase-->AKT-->GSK-3beta-->cyclin D1-->pRB pathway to block uterine epithelial cell proliferation. Mol Endocrinol. 2005;19:1978–90.PubMed
96.
Zurück zum Zitat Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, et al. Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol. 2001;3:1009–13.PubMed Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, et al. Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways. Nat Cell Biol. 2001;3:1009–13.PubMed
97.
Zurück zum Zitat Bhaskar PT, Hay N. The two TORCs and Akt. Dev Cell. 2007;12:487–502.PubMed Bhaskar PT, Hay N. The two TORCs and Akt. Dev Cell. 2007;12:487–502.PubMed
98.
Zurück zum Zitat Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY, Moffat J, et al. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev Cell. 2006;11:859–71.PubMed Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY, Moffat J, et al. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev Cell. 2006;11:859–71.PubMed
99.
Zurück zum Zitat Hayot M, Rodriguez J, Vernus B, Carnac G, Jean E, Allen D, et al. Myostatin up-regulation is associated with the skeletal muscle response to hypoxic stimuli. Mol Cell Endocrinol. 2011;332:38–47.PubMed Hayot M, Rodriguez J, Vernus B, Carnac G, Jean E, Allen D, et al. Myostatin up-regulation is associated with the skeletal muscle response to hypoxic stimuli. Mol Cell Endocrinol. 2011;332:38–47.PubMed
100.
Zurück zum Zitat Favier FB, Costes F, Defour A, Bonnefoy R, Lefai E, Bauge S, et al. Downregulation of Akt/mammalian target of rapamycin pathway in skeletal muscle is associated with increased REDD1 expression in response to chronic hypoxia. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1659–66.PubMed Favier FB, Costes F, Defour A, Bonnefoy R, Lefai E, Bauge S, et al. Downregulation of Akt/mammalian target of rapamycin pathway in skeletal muscle is associated with increased REDD1 expression in response to chronic hypoxia. Am J Physiol Regul Integr Comp Physiol. 2010;298:R1659–66.PubMed
101.
Zurück zum Zitat Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009;19:128–39.PubMed Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009;19:128–39.PubMed
102.
Zurück zum Zitat Springer J, Adams V, Anker SD. Myostatin: regulator of muscle wasting in heart failure and treatment target for cardiac cachexia. Circulation. 2010;121:354–6.PubMed Springer J, Adams V, Anker SD. Myostatin: regulator of muscle wasting in heart failure and treatment target for cardiac cachexia. Circulation. 2010;121:354–6.PubMed
103.
Zurück zum Zitat von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. J Cachexia Sarcopenia Muscle. 2010;1:7–8. von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. J Cachexia Sarcopenia Muscle. 2010;1:7–8.
Metadaten
Titel
The role of myostatin in muscle wasting: an overview
verfasst von
Yulia Elkina
Stephan von Haehling
Stefan D. Anker
Jochen Springer
Publikationsdatum
01.09.2011
Verlag
Springer-Verlag
Erschienen in
Journal of Cachexia, Sarcopenia and Muscle / Ausgabe 3/2011
Print ISSN: 2190-5991
Elektronische ISSN: 2190-6009
DOI
https://doi.org/10.1007/s13539-011-0035-5

Weitere Artikel der Ausgabe 3/2011

Journal of Cachexia, Sarcopenia and Muscle 3/2011 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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