Muscle wasting remains a cause of morbidity and mortality after trauma, burns and infection. Although the mechanisms for the atrophic response are multifactorial and poorly defined, our work documents the causal relationship between the up regulation of inflammatory modulators, particularly TNF"""""""" and NO, and the decreased translational control of muscle protein synthesis. The sepsis-induced defect is manifested not only under basal conditions but also as a reduced responsiveness to leucine which acts as an anabolic nutrient signal. Our data suggest these sepsis-induced changes are mediated by inhibition of mTOR kinase signaling in skeletal muscle. Our long-term goal is to elucidate the cellular and molecular mechanisms by which sepsis impairs muscle protein balance leading to the development of muscle atrophy. To address the questions implicit in this goal our proposed research has the following specific aims: (1) Elucidate the mechanism by which sepsis impairs mTOR kinase activity and cap-dependent translation initiation in muscle under basal conditions and in response to leucine by determining whether sepsis alters the interaction of 4E-BP1 and S6K1 with the scaffold protein raptor, the binding activity of the eIF3 scaffolding complex, and/or the binding of PDCD4 to eIF4A and thereby altering helicase activity. (2) Delineate the mechanism by which sepsis impairs the topology of protein synthesis by altering the intracellular localization of components of the translational apparatus. Specifically, we will assess the role of Vps34, a nutrient-regulated lipid kinase, and Rab5 in regulating intracellular mTOR localization. (3) Determine whether sepsis decreases mTOR kinase activity by altering the phosphorylation and/or binding of the Akt substrate PRAS40. (4) Determine the mechanism by which sepsis-induced changes in microRNAs, particularly miR133, decrease global translation initiation. (5) Elucidate the relative contribution of systemic versus locally produced TNF"""""""" and NO in disrupting mTOR signaling in muscle.
This final aim directly addresses the importance of autocrine/paracrine production of inflammatory mediators in sepsis-induced muscle wasting by using primary cultures of myocytes isolated from TNFR1 null mice or in complementary in vivo studies where shRNA specific for TNF"""""""" is electroporated locally into muscle. Collectively, these studies are unique as they integrate in vitro and in vivo approaches permitting detailed effector mechanisms to be defined and the physiological relevance of the observations to be confirmed. These studies will provide new insights into this critical regulatory system and the integrative control mechanisms exerted by a key nutrient signal. They will lead not only to a thorough understanding of sepsis- induced myopathy but also of other health-related conditions characterized by muscle wasting and nutrient resistance. The proposed studies have a translational underpinning in that they will stimulate the development of novel clinical interventions designed to abrogate the long-term consequences of sepsis- and trauma-induced myopathy which impedes recovery and rehabilitation.

Public Health Relevance

The loss of muscle in patients after infection or traumatic injury delays recovery and may decrease survival. The reason why this wasting occurs is not fully understood, but it cannot be fully corrected by providing adequate nutrition. Our research will determine the mechanism for this muscle atrophy which will ultimately lead to development of novel therapeutic treatment and improve patient outcome.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM038032-25
Application #
8069985
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Dunsmore, Sarah
Project Start
1987-04-01
Project End
2012-05-31
Budget Start
2011-06-01
Budget End
2012-05-31
Support Year
25
Fiscal Year
2011
Total Cost
$326,211
Indirect Cost
Name
Pennsylvania State University
Department
Physiology
Type
Schools of Medicine
DUNS #
129348186
City
Hershey
State
PA
Country
United States
Zip Code
17033
Crowell, Kristen T; Moreno, Samantha; Steiner, Jennifer L et al. (2018) Temporally Distinct Regulation of Pathways Contributing to Cardiac Proteostasis During the Acute and Recovery Phases of Sepsis. Shock 50:616-626
Crowell, Kristen T; Soybel, David I; Lang, Charles H (2017) Inability to replete white adipose tissue during recovery phase of sepsis is associated with increased autophagy, apoptosis, and proteasome activity. Am J Physiol Regul Integr Comp Physiol 312:R388-R399
Crowell, Kristen T; Phillips, Brett E; Kelleher, Shannon L et al. (2017) Immune and metabolic responses in early and late sepsis during mild dietary zinc restriction. J Surg Res 210:47-58
Crowell, Kristen T; Soybel, David I; Lang, Charles H (2017) Restorative Mechanisms Regulating Protein Balance in Skeletal Muscle During Recovery From Sepsis. Shock 47:463-473
Crowell, Kristen T; Kelleher, Shannon L; Soybel, David I et al. (2016) Marginal dietary zinc deprivation augments sepsis-induced alterations in skeletal muscle TNF-? but not protein synthesis. Physiol Rep 4:
Atherton, Philip J; Greenhaff, Paul L; Phillips, Stuart M et al. (2016) Control of skeletal muscle atrophy in response to disuse: clinical/preclinical contentions and fallacies of evidence. Am J Physiol Endocrinol Metab 311:E594-604
Gordon, Bradley S; Steiner, Jennifer L; Williamson, David L et al. (2016) Emerging role for regulated in development and DNA damage 1 (REDD1) in the regulation of skeletal muscle metabolism. Am J Physiol Endocrinol Metab 311:E157-74
Steiner, Jennifer L; Crowell, Kristen T; Kimball, Scot R et al. (2015) Disruption of REDD1 gene ameliorates sepsis-induced decrease in mTORC1 signaling but has divergent effects on proteolytic signaling in skeletal muscle. Am J Physiol Endocrinol Metab 309:E981-94
Gordon, Bradley S; Williamson, David L; Lang, Charles H et al. (2015) Nutrient-induced stimulation of protein synthesis in mouse skeletal muscle is limited by the mTORC1 repressor REDD1. J Nutr 145:708-13
Steiner, Jennifer L; Lang, Charles H (2015) Sepsis attenuates the anabolic response to skeletal muscle contraction. Shock 43:344-51

Showing the most recent 10 out of 41 publications