Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Troy A. Hornberger is active.

Publication


Featured researches published by Troy A. Hornberger.


Biochemical Journal | 2004

Mechanical stimuli regulate rapamycin-sensitive signalling by a phosphoinositide 3-kinase-, protein kinase B- and growth factor-independent mechanism

Troy A. Hornberger; Rudy Stuppard; Kevin E. Conley; Mark J. Fedele; Marta L. Fiorotto; Eva R. Chin; Karyn A. Esser

In response to growth factors, mTOR (mammalian target of rapamycin) has been identified as a central component of the signalling pathways that control the translational machinery and cell growth. Signalling through mTOR has also been shown to be necessary for the mechanical load-induced growth of cardiac and skeletal muscles. Although the mechanisms involved for mechanically induced activation of mTOR are not known, it has been suggested that activation of PI3K (phosphoinositide 3-kinase) and protein kinase B (Akt), via the release of locally acting growth factors, underlies this process. In the present study, we show that mechanically stimulating (passive stretch) the skeletal muscle ex vivo results in the activation of mTOR-dependent signalling events. The activation of mTOR-dependent signalling events was necessary for an increase in translational efficiency, demonstrating the physiological significance of this pathway. Using pharmacological inhibitors, we show that activation of mTOR-dependent signalling occurs through a PI3K-independent pathway. Consistent with these results, mechanically induced signalling through mTOR was not disrupted in muscles from Akt1-/- mice. In addition, ex vivo co-incubation experiments, along with in vitro conditioned-media experiments, demonstrate that a mechanically induced release of locally acting autocrine/paracrine growth factors was not sufficient for the activation of the mTOR pathway. Taken together, our results demonstrate that mechanical stimuli can activate the mTOR pathway independent of PI3K/Akt1 and locally acting growth factors. Thus mechanical stimuli and growth factors provide distinct inputs through which mTOR co-ordinates an increase in the translational efficiency.


The FASEB Journal | 2011

Novel insights into the regulation of skeletal muscle protein synthesis as revealed by a new nonradioactive in vivo technique

Craig A. Goodman; Danielle M. Mabrey; John W. Frey; Man Hing Miu; Enrico K. Schmidt; Philippe Pierre; Troy A. Hornberger

In this study, the principles of surface sensing of translation (SUnSET) were used to develop a nonradioactive method for ex vivo and in vivo measurements of protein synthesis (PS). Compared with controls, we first demonstrate excellent agreement between SUnSET and a [3H]phenylalanine method when detecting synergist ablation‐induced increases in skeletal muscle PS ex vivo. We then show that SUnSET can detect the same synergist ablation‐induced increase in PS when used in vivo (IV‐SUnSET). In addition, IV‐SUnSET detected food deprivation‐induced decreases in PS in the heart, kidney, and skeletal muscles, with similar changes being visualized with an immunohistochemical version of IV‐SUnSET (IV‐IHC‐SUnSET). By combining IV‐IHC‐SUnSET with in vivo transfection, we demonstrate that constitutively active PKB induces a robust increase in skeletal muscle PS. Furthermore, transfection with Ras homolog enriched in brain (Rheb) revealed that a PKB‐independent activation of mammalian target of rapamycin is also sufficient to induce an increase in skeletal muscle PS. Finally, IV‐IHC‐SUnSET exposed the existence of fiber type‐dependent differences in skeletal muscle PS, with PS in type 2B and 2X fibers being significantly lower than that in type 2A fibers within the same muscle. Thus, our nonradioactive method allowed us to accurately visualize and quantify PS under various ex vivo and in vivo conditions and revealed novel insights into the regulation of PS in skeletal muscle.—Goodman, C. A., Mabrey, D. M., Frey, J. W., Miu, M. H., Schmidt, E. K., Pierre, P., Hornberger, T. A. Novel insights into the regulation of skeletal muscle protein synthesis as revealed by a new nonradioactive in vivo technique. FASEB J. 25, 1028–1039 (2011). www.fasebj.org


Proceedings of the National Academy of Sciences of the United States of America | 2010

CLOCK and BMAL1 regulate MyoD and are necessary for maintenance of skeletal muscle phenotype and function

Jessica L. Andrews; Xiping Zhang; John J. McCarthy; Erin L. McDearmon; Troy A. Hornberger; Brenda Russell; Kenneth S. Campbell; Sandrine Arbogast; Michael B. Reid; John R. Walker; John B. Hogenesch; Joseph S. Takahashi; Karyn A. Esser

MyoD, a master regulator of myogenesis, exhibits a circadian rhythm in its mRNA and protein levels, suggesting a possible role in the daily maintenance of muscle phenotype and function. We report that MyoD is a direct target of the circadian transcriptional activators CLOCK and BMAL1, which bind in a rhythmic manner to the core enhancer of the MyoD promoter. Skeletal muscle of ClockΔ19 and Bmal1−/− mutant mice exhibited ∼30% reductions in normalized maximal force. A similar reduction in force was observed at the single-fiber level. Electron microscopy (EM) showed that the myofilament architecture was disrupted in skeletal muscle of ClockΔ19, Bmal1−/−, and MyoD−/− mice. The alteration in myofilament organization was associated with decreased expression of actin, myosins, titin, and several MyoD target genes. EM analysis also demonstrated that muscle from both ClockΔ19 and Bmal1−/− mice had a 40% reduction in mitochondrial volume. The remaining mitochondria in these mutant mice displayed aberrant morphology and increased uncoupling of respiration. This mitochondrial pathology was not seen in muscle of MyoD−/− mice. We suggest that altered expression of both Pgc-1α and Pgc-1β in ClockΔ19 and Bmal1−/− mice may underlie this pathology. Taken together, our results demonstrate that disruption of CLOCK or BMAL1 leads to structural and functional alterations at the cellular level in skeletal muscle. The identification of MyoD as a clock-controlled gene provides a mechanism by which the circadian clock may generate a muscle-specific circadian transcriptome in an adaptive role for the daily maintenance of adult skeletal muscle.


The Journal of Physiology | 2009

The role of phosphoinositide 3-kinase and phosphatidic acid in the regulation of mammalian target of rapamycin following eccentric contractions

T. K. O’Neil; L. R. Duffy; John W. Frey; Troy A. Hornberger

Resistance exercise induces a hypertrophic response in skeletal muscle and recent studies have begun to shed light on the molecular mechanisms involved in this process. For example, several studies indicate that signalling by the mammalian target of rapamycin (mTOR) is necessary for a hypertrophic response. Furthermore, resistance exercise has been proposed to activate mTOR signalling through an upstream pathway involving the phosphoinositide 3‐kinase (PI3K) and protein kinase B (PKB); however, this hypothesis has not been thoroughly tested. To test this hypothesis, we first evaluated the temporal pattern of signalling through PI3K–PKB and mTOR following a bout of resistance exercise with eccentric contractions (EC). Our results indicated that the activation of signalling through PI3K–PKB is a transient event (<15 min), while the activation of mTOR is sustained for a long duration (>12 h). Furthermore, inhibition of PI3K–PKB activity did not prevent the activation of mTOR signalling by ECs, indicating that PI3K–PKB is not part of the upstream regulatory pathway. These observations led us to investigate an alternative pathway for the activation of mTOR signalling involving the synthesis of phosphatidic acid (PA) by phospholipase D (PLD). Our results demonstrate that ECs induce a sustained elevation in [PA] and inhibiting the synthesis of PA by PLD prevented the activation of mTOR. Furthermore, we determined that similar to ECs, PA activates mTOR signalling through a PI3K–PKB‐independent mechanism. Combined, the results of this study indicate that the activation of mTOR following eccentric contractions occurs through a PI3K–PKB‐independent mechanism that requires PLD and PA.


The Journal of Physiology | 2011

The role of skeletal muscle mTOR in the regulation of mechanical load-induced growth

Craig A. Goodman; John W. Frey; Danielle M. Mabrey; Brittany L. Jacobs; Hannah C. Lincoln; Jae-Sung You; Troy A. Hornberger

Non‐Technical Summary  Chronic mechanical loading (CML) of skeletal muscle induces growth and this effect can be blocked by the drug rapamycin. Rapamycin is considered to be a highly specific inhibitor of the mammalian target of rapamycin (mTOR), and thus, many have concluded that mTOR plays a key role in CML‐induced growth. However, direct evidence that mTOR confers the CML‐induced activation of growth promoting events such as hypertrophy, hyperplasia and ribosome biogenesis is lacking. This study addressed that gap in knowledge by using a specialized line of transgenic mice. Surprisingly, the results indicate that only a few of the growth promoting events induced by CML are fully dependent on mTOR signalling (e.g. hypertrophy). These results advance our understanding of the molecular mechanisms that regulate skeletal muscle mass and should help future studies aimed at identifying targets for therapies that can prevent the loss of muscle mass during conditions such as bedrest, immobilization, and ageing.


Cellular Signalling | 2011

Recent progress toward understanding the molecular mechanisms that regulate skeletal muscle mass.

Craig A. Goodman; David L. Mayhew; Troy A. Hornberger

The maintenance of muscle mass is critical for health and issues associated with the quality of life. Over the last decade, extensive progress has been made with regard to our understanding of the molecules that regulate skeletal muscle mass. Not surprisingly, many of these molecules are intimately involved in the regulation of protein synthesis and protein degradation [e.g. the mammalian target of rapamycin (mTOR), eukaryotic initiation factor 2B (eIF2B), eukaryotic initiation factor 3f (eIF3f) and the forkhead box O (FoxO) transcription factors]. It is also becoming apparent that molecules which sense, or control, the energetic status of the cell play a key role in the regulation of muscle mass [e.g. AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator-1 α (PGC1α)]. In this review we will attempt to summarize the current knowledge of how these molecules regulate skeletal muscle mass.


Molecular Biology of the Cell | 2010

A Phosphatidylinositol 3-Kinase/Protein Kinase B-independent Activation of Mammalian Target of Rapamycin Signaling Is Sufficient to Induce Skeletal Muscle Hypertrophy

Craig A. Goodman; Man Hing Miu; John W. Frey; Danielle M. Mabrey; Hannah C. Lincoln; Yejing Ge; Jie Chen; Troy A. Hornberger

Overexpression of Rheb activates mTOR signaling via a PI3K/PKB-independent mechanism and is sufficient to induce skeletal muscle hypertrophy. The hypertrophic effects of Rheb are driven through a rapamycin-sensitive (RS) mechanism, mTOR is the RS element that confers the hypertrophy and the kinase activity of mTOR is necessary for this event.


The International Journal of Biochemistry & Cell Biology | 2011

Mechanotransduction and the Regulation of mTORC1 Signaling in Skeletal Muscle

Troy A. Hornberger

Mechanical stimuli play a major role in the regulation of skeletal muscle mass, and the maintenance of muscle mass contributes significantly to disease prevention and issues associated with the quality of life. Although the link between mechanical signals and the regulation of muscle mass has been recognized for decades, the mechanisms involved in converting mechanical information into the molecular events that control this process remain poorly defined. Nevertheless, our knowledge of these mechanisms is advancing and recent studies have revealed that signaling through a protein kinase called the mammalian target of rapamycin (mTOR) plays a central role in this event. In this review we will, (1) discuss the evidence which implicates mTOR in the mechanical regulation of skeletal muscle mass, (2) provide an overview of the mechanisms through which signaling by mTOR can be regulated, and (3) summarize our current knowledge of the potential mechanisms involved in the mechanical activation of mTOR signaling.


Journal of Cellular Biochemistry | 2006

Mechanical stimuli and nutrients regulate rapamycin‐sensitive signaling through distinct mechanisms in skeletal muscle

Troy A. Hornberger; Shu Chien

The mammalian target of rapamycin (mTOR) has been identified as a growth factor and nutrient‐sensitive molecule that controls the translational machinery and cell growth. Rapamycin‐sensitive (RS) signaling events have also been shown to be necessary for mechanical load‐induced growth of skeletal muscle, but the mechanisms involved in the mechanical activation of RS signaling are not known. The finding that mechanical stimuli induce nutrient uptake in skeletal muscle raises the possibility that mechanically induced RS signaling is mediated via a nutrient‐dependent mechanism. To investigate this hypothesis, skeletal muscles (ex vivo) were stimulated with nutrients or intermittent mechanical stretch and the phosphorylation of p70S6k [P‐p70(389)], PKB [P‐PKB], mTOR [P‐mTOR(2481)], and p38 [P‐p38] was assessed. In comparison to vehicle‐treated controls, both nutrient and mechanical stimuli induced P‐p70(389), neither stimulus altered P‐PKB or P‐mTOR(2481), and only mechanical stimuli induced P‐p38. The nutrient and mechanically induced increase in P‐p70(389) was blocked by rapamycin, but only nutrient‐induced signaling to P‐p70(389) was blocked by wortmannin. Furthermore, the mechanically induced increase in P‐p70(389) was not impaired by the removal of exogenous nutrients. Taken together, these results indicate that exogenous nutrients are not required for mechanically induced RS signaling and that nutrient and mechanical stimuli activate RS signaling through distinct upstream mechanisms. J. Cell. Biochem. 97: 1207–1216, 2006.


Molecular Biology of the Cell | 2010

A PI3K/PKB-Independent Activation of mTOR Signaling Is Sufficient to Induce Skeletal Muscle Hypertrophy

Craig A. Goodman; Man Hing Miu; John W. Frey; Danielle M. Mabrey; Hannah C. Lincoln; Yejing Ge; Jie Chen; Troy A. Hornberger

Overexpression of Rheb activates mTOR signaling via a PI3K/PKB-independent mechanism and is sufficient to induce skeletal muscle hypertrophy. The hypertrophic effects of Rheb are driven through a rapamycin-sensitive (RS) mechanism, mTOR is the RS element that confers the hypertrophy and the kinase activity of mTOR is necessary for this event.

Collaboration


Dive into the Troy A. Hornberger's collaboration.

Top Co-Authors

Avatar

Craig A. Goodman

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

John W. Frey

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jae-Sung You

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hannah C. Lincoln

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Brittany L. Jacobs

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Danielle M. Mabrey

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Man Hing Miu

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Rachel M. McNally

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Shu Chien

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge