ArticlePDF Available

Improving Type 2 Diabetes Through a Distinct Adrenergic Signaling Pathway Involving mTORC2 That Mediates Glucose Uptake in Skeletal Muscle

Authors:

Abstract

Type 2 diabetes is an increasing worldwide epidemic that poses major health problems. We have identified a novel physiological system that increases glucose uptake in skeletal muscle but not in white adipocytes. Activation of this system improves glucose tolerance in Goto-Kakizaki rats or mice fed a high fat diet, which are established models for type 2 diabetes. The pathway involves activation of β2-adrenoceptors that increase cAMP levels and activate PKA that phosphorylates mammalian target of rapamycin complex 2 (mTORC2) at S2481. The active mTORC2 causes translocation of GLUT4 to the plasma membrane and glucose uptake without the involvement of Akt or AS160. Stimulation of glucose uptake into skeletal muscle following activation of the sympathetic nervous system is likely to be of high physiological relevance since mTOR complex 2 activation was observed at the cellular, tissue and whole animal level in rodent and human systems. This signaling pathway provides new opportunities for the treatment of type 2 diabetes.
Masaaki Sato,
1,2,3
Nodi Dehvari,
1
Anette I. Öberg,
2,3
Roger J. Summers,
2,3
Dana S. Hutchinson,
2,3
and Tore Bengtsson
1
RESPONSE TO COMMENT ON SATO ET AL.
Improving Type 2 Diabetes
Through a Distinct Adrenergic
Signaling Pathway Involving
mTORC2 That Mediates Glucose
Uptake in Skeletal Muscle.
Diabetes 2014;63:41154129
Diabetes 2014;63:e22e23 | DOI: 10.2337/db14-1283
We thank Xiang et al. (1) for their comments on our article
published in Diabetes (2) that shows for the rst time that
activation of b
2
-adrenoceptors promotes GLUT4-translocation
and glucose uptake in skeletal muscle via a specic pathway
dependent on mammalian target of rapamycin (mTOR) com-
plex 2 (mTORC2). Because this pathway bypasses several of
the proteins used in the insulin pathway, which are down-
regulated and desensitized in type 2 diabetes, we hypothesize
that this pathway may be used to regulate glucose uptake and
blood glucose in diabetic patients.
However, Xiang et al. (1) raise important concerns re-
garding the use of b
2
-adrenoceptor agonists to treat type
2 diabetes. As they point out, systemic activation of b
2
-
adrenoceptors can lead to an acute rise in blood glucose
levels by increasing hepatic glucose output, and this could
be detrimental for diabetic patients. Stimulation of b
2
-
adrenoceptors can regulate both glucose output from the
liver and its uptake into skeletal muscle. However, our
novel nding makes it feasible to develop compounds
that specically enhance glucose uptake into skeletal mus-
cle without activating glucose output from the liver.
There are several ways in which the b
2
-adrenergic pathway
could be used to develop new medicines to treat type 2 di-
abetes. An emerging paradigm in pharmacology is the recog-
nition of biased ligands that can stimulate a preferred subset
of signaling pathways activated by the receptor (3). Another
possibility would be to directly activate specicproteinsinthe
novel b
2
-adrenergic pathway. Both approaches could pro-
mote glucose uptake in skeletal muscle without accompany-
ing side effects in the liver and other tissues.
Furthermore, the rise in blood glucose after sympathetic
stimulation is a short-term effect, as illustrated in another
article by Xiang et al. (4) in which orthopedic trauma in-
duced an increase in blood glucose that peaked after 2 h. In
contrast, the long-term effect of b
2
-adrenoceptor agonist
treatment is an improvement of glucose tolerance (2),
suggesting that glucose uptake into skeletal muscle out-
weighs glucose release from the liver.
Anal concern raised by Xiang et al. (1) is that our study
does not directly show that the mTORC2 pathway is involved
in the improvement of glucose tolerance in type 2 diabetes.
Our study clearly shows that there is a novel b
2
-adrenoceptor
signaling pathway that involves mTORC2 and promotes glu-
cose uptake in skeletal muscle. This was shown in vitro, ex
vivo, and in vivo. It is thus very likely that the novel pathway
also mediates some or all of the benecial effects and im-
provement of glucose tolerance induced by b
2
-adrenoceptor
stimulation in type 2 diabetes, although further studies are
required to fully understand this mechanism. These studies
should dissect different parts of the physiological response,
focusing particularly on glucose uptake in skeletal muscle,
to better understand how this can best be used for the de-
velopment of new medicines. Although b
2
-adrenoceptor
stimulation of glucose uptake in skeletal muscle has been
1
Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm
University, Stockholm, Sweden
2
Department of Pharmacology, Monash University, Clayton, Victoria, Australia
3
Drug Discovery Biology Laboratory, Monash Institute of Pharmaceutical Sciences,
Monash University, Parkville, Victoria, Australia
Corresponding author: Tore Bengtsson, tore.bengtsson@su.se.
M.S., N.D., and A.I.Ö. contributed equally to the response.
© 2014 by the American Diabetes Association. Readers may use this article as
long as the work is properly cited, the use is educational and not for prot, and
the work is not altered.
e22 Diabetes Volume 63, December 2014
e-LETTERS COMMENTS AND RESPONSES
a neglected area of research, we believe that our study clearly
identies novel targets that can be exploited in the search
for new treatments for type 2 diabetes.
Duality of Interest. No potential conicts of interest relevant to this article
were reported.
References
1. Xiang L, Mittwede PN, Hester RL. Comment on Sato et al. Improving type 2
diabetes through a distinct adrenergic signaling pathway involving mTORC2 that
mediates glucose uptake in skeletal muscle. Diabetes 2014;63:41154129 (Letter).
Diabetes 2014;63:e20. DOI: 10.2337/db14-1187
2. Sato M, Dehvari N, Öberg AI, et al. Improving type 2 diabetes through
a distinct adrenergic signaling pathway involving mTORC2 that mediates glucose
uptake in skeletal muscle. Diabetes 2014;63:41154129
3. Evans BA, Sato M, Sarwar M, Hutchinson DS, Summers RJ. Ligand-directed
signalling at b-adrenoceptors. Br J Pharmacol 2010;159:10221038
4. Xiang L, Lu S, Mittwede PN, Clemmer JS, Husband GW, Hester RL. b
2
adrenoreceptor blockade improves early post-trauma hyperglycemia and
pulmonary injury in obese rats. Am J Physiol Heart Circ Physiol 2014;307:
H621H627.
diabetes.diabetesjournals.org Response e23
... One such response, β 2 -AR mediated glucose uptake in skeletal muscle, is attenuated by inhibition of PKA/AC and mimicked by cyclic adenosine monophosphate (cAMP) analogs. [2][3][4] In skeletal muscle, cAMP levels rise rapidly in response to high efficacy β 2 -AR agonists such as isoprenaline 5 before cAMP is degraded by phosphodiesterases, whereas β 2 -AR mediated glucose uptake by isoprenaline increases more slowly and the effect is sustained for up to 20 h. 5 This suggests that the transient nature of the β 2 -AR cAMP response may occur in parallel with an alternative mechanism that maintains increased glucose uptake, highlighting the importance of understanding the molecular mechanisms involved. ...
... Transcriptional regulation may also be a factor considering the differences in time courses observed but isoprenaline-mediated glucose uptake in skeletal muscle is independent of both transcription and translation, at least 2 h post-receptor stimulation. 4,5 In addition, partial agonists such as BRL37344 produce only modest cAMP responses yet have similar efficacy to isoprenaline for glucose uptake. ...
... Our previous work has involved investigating the mechanism whereby β 2 -ARs increase skeletal muscle glucose uptake. [2][3][4][5]22 We and others have shown that activation of β 2 -ARs increases glucose uptake in skeletal muscle using cell lines (mouse C 2 C 12 , rat L6 skeletal muscle, human skeletal muscle cells) and isolated skeletal muscle strips, and that β 2 -AR agonist administration in vivo improves glucose tolerance in diabetic rats and obese mice. 4 A level of complexity arises from in vivo studies in rodents using BRL37344 as it is a dual rodent β 2 −/β 3 -AR agonist and its beneficial effects at least in rodents are most likely due to its action at adipocyte β 3 -ARs and skeletal muscle β 2 -ARs. ...
Article
Full-text available
Truncation of the C‐terminal tail of the β 2 ‐AR, transfection of βARKct or over‐expression of a kinase‐dead GRK mutant reduces isoprenaline‐stimulated glucose uptake, indicating that GRK is important for this response. We explored whether phosphorylation of the β 2 ‐AR by GRK2 has a role in glucose uptake or if this response is related to the role of GRK2 as a scaffolding protein. CHO‐GLUT4myc cells expressing wild‐type and mutant β 2 ‐ARs were generated and receptor affinity for [ ³ H]‐CGP12177A and density of binding sites determined together with the affinity of isoprenaline and BRL37344. Following receptor activation by β 2 ‐AR agonists, cAMP accumulation, GLUT4 translocation, [ ³ H]‐2‐deoxyglucose uptake, and β 2 ‐AR internalization were measured. Bioluminescence resonance energy transfer was used to investigate interactions between β 2 ‐AR and β‐arrestin2 or between β 2 ‐AR and GRK2. Glucose uptake after siRNA knockdown or GRK inhibitors was measured in response to β 2 ‐AR agonists. BRL37344 was a poor partial agonist for cAMP generation but displayed similar potency and efficacy to isoprenaline for glucose uptake and GLUT4 translocation. These responses to β 2 ‐AR agonists occurred in CHO‐GLUT4myc cells expressing β 2 ‐ARs lacking GRK or GRK/PKA phosphorylation sites as well as in cells expressing the wild‐type β 2 ‐AR. However, β 2 ‐ARs lacking phosphorylation sites failed to recruit β‐arrestin2 and did not internalize. GRK2 knock‐down or GRK2 inhibitors decreased isoprenaline‐stimulated glucose uptake in rat L6 skeletal muscle cells. Thus, GRK phosphorylation of the β 2 ‐AR is not associated with isoprenaline‐ or BRL37344‐stimulated glucose uptake. However, GRKs acting as scaffold proteins are important for glucose uptake as GRK2 knock‐down or GRK2 inhibition reduces isoprenaline‐stimulated glucose uptake.
... mTORC2 will then on its turn phosphorylate AKT at Ser473, resulting in full AKT activation [63,64]. Additional stimuli that can trigger mTORC2 activation include adrenergic signaling via G-protein coupled receptors (GPCR), such as the β2-adrenoceptor, which stimulates cAMP accumulation and activation of cAMP-dependent protein kinase (PKA), leading to phosphorylation of mTORC2 [65]. Also, AMPK appears to be sufficient to increase mTORC2 catalytic activity towards AKT in an mTORC1-independent manner [66]. ...
Article
Full-text available
Multiple Myeloma (MM), a cancer of terminally differentiated plasma cells, is the second most prevalent hematological malignancy and is incurable due to the inevitable development of drug resistance. Intense protein synthesis is a distinctive trait of MM cells, supporting the massive production of clonal immunoglobulins or free light chains. The mammalian target of rapamycin (mTOR) kinase is appreciated as a master regulator of vital cellular processes, including regulation of metabolism and protein synthesis, and can be found in two multiprotein complexes, mTORC1 and mTORC2. Dysregulation of these complexes is implicated in several types of cancer, including MM. Since mTOR has been shown to be aberrantly activated in a large portion of MM patients and to play a role in stimulating MM cell survival and resistance to several existing therapies, understanding the regulation and functions of the mTOR complexes is vital for the development of more effective therapeutic strategies. This review provides a general overview of the mTOR pathway, discussing key discoveries and recent insights related to the structure and regulation of mTOR complexes. Additionally, we highlight findings on the mechanisms by which mTOR is involved in protein synthesis and delve into mTOR-mediated processes occurring in MM. Finally, we summarize the progress and current challenges of drugs targeting mTOR complexes in MM.
... The activation of cAMP signaling pathway is closely related to increased glucose uptake in skeletal muscle. This physiological process is independent of insulin regulation, and the regulation of cAMP signaling pathway in skeletal muscle can be used as an alternative pathway for glucose processing and blood sugar control [30]. ...
Article
Full-text available
Objective There is increasing evidence that type 2 diabetes mellitus (T2DM) is an independent risk factor for the occur of tendinopathy. Therefore, this study is the first to explore the dynamic changes of the “gene profile” of supraspinatus tendon in rats at different time points after T2DM induction through transcriptomics, providing potential molecular markers for exploring the pathogenesis of diabetic tendinopathy. Methods A total of 40 Sprague-Dawley rats were randomly divided into normal (NG, n = 10) and T2DM groups (T2DM, n = 30) and subdivided into three groups according to the duration of diabetes: T2DM-4w, T2DM-8w, and T2DM-12w groups; the duration was calculated from the time point of T2DM rat model establishment. The three comparison groups were set up in this study, T2DM-4w group vs. NG, T2DM-8w group vs. NG, and T2DM-12w group vs. NG. Differentially expressed genes (DEGs) in 3 comparison groups were screened. The intersection of the three comparison groups’ DEGs was defined as key genes that changed consistently in the supraspinatus tendon after diabetes induction. Cluster analysis, gene ontology (GO) functional annotation analysis and Kyoto encyclopedia of genes and genomes (KEGG) functional annotation and enrichment analysis were performed for DEGs. Results T2DM-4w group vs. NG, T2DM-8w group vs. NG, and T2DM-12w group vs. NG detected 519 (251 up-regulated and 268 down-regulated), 459 (342 up-regulated and 117 down-regulated) and 328 (255 up-regulated and 73 down-regulated) DEGs, respectively. 103 key genes of sustained changes in the supraspinatus tendon following induction of diabetes, which are the first identified biomarkers of the supraspinatus tendon as it progresses through the course of diabetes.The GO analysis results showed that the most significant enrichment in biological processes was calcium ion transmembrane import into cytosol (3 DEGs). The most significant enrichment in cellular component was extracellular matrix (9 DEGs). The most significant enrichment in molecular function was glutamate-gated calcium ion channel activity (3 DEGs). The results of KEGG pathway enrichment analysis showed that there were 17 major pathways (p < 0.05) that diabetes affected supratinusculus tendinopathy, including cAMP signaling pathway and Calcium signaling pathway. Conclusions Transcriptomics reveals dynamic changes in the“gene profiles”of rat supraspinatus tendon at three different time points after diabetes induction. The 103 DEGs identified in this study may provide potential molecular markers for exploring the pathogenesis of diabetic tendinopathy, and the 17 major pathways enriched in KEGG may provide new ideas for exploring the pathogenesis of diabetic tendinopathy.
... The β2 adrenergic receptors are involved in glucose uptake in both in vitro myotubes and the skeletal muscle of a healthy subject [14]. The ablation of β2 adrenergic receptors in mice results in hyperglycemia, but the supplementation of obese mice or the rat model for human type 2 diabetes mellitus by the β2 adrenergic agonists improves glucose tolerance in these rodents [14][15][16]. The state of reduction in the density of functional β2 receptors in preeclamptic women may have an additional consequence in maternal response to the other stimulators of these receptors, including the CgA and its derivative peptides such as CST [17]. ...
Article
Full-text available
Preeclampsia (PE) is a unique pregnancy disorder affecting women across the world. It is characterized by the new onset of hypertension with coexisting end-organ damage. Although the disease has been known for centuries, its exact pathophysiology and, most importantly, its prevention remain elusive. The basis of its associated molecular changes has been attributed to the placenta and the hormones regulating its function. One such hormone is chromogranin A (CgA). In the placenta, CgA is cleaved to form a variety of biologically active peptides, including catestatin (CST), known inter alia for its vasodilatory effects. Recent studies indicate that the CST protein level is diminished both in patients with hypertension and those with PE. Therefore, the aim of the present paper is to review the most recent and most relevant in vitro, in vivo, and clinical studies to provide an overview of the proposed impact of CST on the molecular processes of PE and to consider the possibilities for future experiments in this area.
Chapter
The sympathetic nervous system plays an important role in the regulation of endocrine pancreatic function, most importantly insulin release. Among the nine adrenoceptor (AR) subtypes, the α2A-AR appears to be the subtype most abundantly expressed in the human pancreas. While α2- and β-AR have opposing effects, the net response to sympathetic stimulation is inhibition of insulin secretion mediated by α2-AR located in the plasma membrane of pancreatic β cells. This inhibition may be present physiologically as evidenced by increased insulin secretion in healthy and diabetic humans and animals in response to α2-AR antagonists, a finding that was confirmed in all studies. Based on such data and on an association of an α2A-AR polymorphism, that increases receptor expression levels, with an elevated risk for diabetes, increased α2A-AR signaling in the pancreatic β cells has been proposed as a risk factor for the development of type 2 diabetes. Thus, the α2A-AR was proposed as a drug target for the treatment of some forms of type 2 diabetes. Drug research and development programs leveraging this mechanism have reached the clinical stage, but none have resulted in an approved medicine due to a limited efficacy. While β-AR agonists can increase circulating insulin levels in vivo, it remains controversial whether this includes a direct effect on β cells or occurs secondary to general metabolic effects. Therefore, the regulation of endocrine pancreatic function is physiologically interesting but may be of limited therapeutic relevance.
Article
Context Given the promising effects of prolonged treatment with beta2-agonist on insulin sensitivity in animals and nondiabetic individuals, the beta2-adrenergic receptor has been proposed as a target to counter peripheral insulin resistance. On the other hand, rodent studies also reveal that beta2-agonists acutely impair insulin action, posing a potential caveat for their use in treating insulin resistance. Objective To assess the impact of beta2-agonist on muscle insulin action and glucose metabolism and identify the underlying mechanism(s) in 10 insulin-resistant subjects. Methods and participants In a crossover design, we assessed the effect of beta2-agonist on insulin-stimulated muscle glucose uptake during a 3-hour hyperinsulinemic isoglycemic clamp with and without intralipid infusion in 10 insulin-resistant, overweight subjects. Two hours into the clamp, we infused beta2-agonist. We collected muscle biopsies before, 2 hours into, and by the end of the clamp and analyzed them using metabolomic and lipidomic techniques. Results We establish that beta2-agonist, independently from and additively to intralipid, impairs insulin-stimulated muscle glucose uptake via different mechanisms. In combination, beta2-agonist and intralipid nearly eliminates insulin-dependent muscle glucose uptake. Although both beta2-agonist and intralipid elevated muscle glucose-6-phosphate, only intralipid caused accumulation of downstream muscle glycolytic intermediates, whereas beta2-agonist attenuated incorporation of glucose into glycogen. Conclusion Our findings suggest that beta2-agonist inhibits glycogenesis, whereas intralipid inhibits glycolysis in skeletal muscle of insulin-resistant individuals. These results should be addressed in future treatment of insulin resistance with beta2-agonist.
Article
Full-text available
The mechanistic target of rapamycin, mTOR, controls cell metabolism in response to growth signals and stress stimuli. The cellular functions of mTOR are mediated by two distinct protein complexes, mTOR complex 1 (mTORC1) and mTORC2. Rapamycin and its analogs are currently used in the clinic to treat a variety of diseases and have been instrumental in delineating the functions of its direct target, mTORC1. Despite the lack of a specific mTORC2 inhibitor, genetic studies that disrupt mTORC2 expression unravel the functions of this more elusive mTOR complex. Like mTORC1 which responds to growth signals, mTORC2 is also activated by anabolic signals but is additionally triggered by stress. mTORC2 mediates signals from growth factor receptors and G-protein coupled receptors. How stress conditions such as nutrient limitation modulate mTORC2 activation to allow metabolic reprogramming and ensure cell survival remains poorly understood. A variety of downstream effectors of mTORC2 have been identified but the most well-characterized mTORC2 substrates include Akt, PKC, and SGK, which are members of the AGC protein kinase family. Here, we review how mTORC2 is regulated by cellular stimuli including how compartmentalization and modulation of complex components affect mTORC2 signaling. We elaborate on how phosphorylation of its substrates, particularly the AGC kinases, mediates its diverse functions in growth, proliferation, survival, and differentiation. We discuss other signaling and metabolic components that cross-talk with mTORC2 and the cellular output of these signals. Lastly, we consider how to more effectively target the mTORC2 pathway to treat diseases that have deregulated mTOR signaling.
Article
Full-text available
Sato et al. (1) showed that β2-adrenoreceptor–mediated phosphorylation of mammalian target of rapamycin complex 2 (mTORC2) in skeletal muscles increased GLUT4 translocation to the plasma membrane and thus increased cellular glucose uptake. In diabetes and obesity, an upregulated G-protein–coupled receptor kinase, which inhibits G-protein–coupled receptors, has been reported to contribute to insulin resistance in skeletal muscle (2). In support of this, the current study demonstrated an increased peripheral glucose uptake following β2-adrenoreceptor activation in diabetic rats and obese mice (1). However, the statement that “this signaling pathway provides new opportunities for the treatment of type 2 diabetes” (1) may be debatable because the impacts of systemic β2-adrenoreceptor activation on glucose control and hemodynamic changes are not considered. Increased sympathetic nervous system activity (exercise and the fight or flight response, as Sato et al. mentioned) is associated not only with increased glucose uptake by skeletal muscle but also with increased hepatic glycogenolysis and/or glucagon release, likely leading to an increase in plasma glucose levels. Plasma glucose and glucagon levels can increase significantly during exercise despite increased glucose uptake in exercising muscle. We recently demonstrated that traumatic injury rapidly increased plasma glucose levels via β2-adrenoreceptor–dependent glycogenolysis in the liver. Treatment with a selective β2-adrenoreceptor antagonist blunted the hepatic glycogenolysis and the increase in plasma glucose levels (3). Thus, following systemic β2-adrenoreceptor activation, the beneficial effect from increased peripheral glucose uptake in skeletal muscle may be overridden by simultaneous increases in hepatic glycogenolysis and/or glucagon release. Notably, due to insulin resistance, obese rats exhibited exacerbated post-trauma hyperglycemia as compared with lean Zucker rats (3). Therefore, the net increase in plasma glucose following β2-adrenoreceptor activation could be more profound in obesity and type 2 diabetes. Sato et al. (1) showed that systemic treatment with β2-adrenoreceptor agonist improved glucose tolerance in diabetic rats and obese mice. However, whether this is due to the same mechanism (mTORC2 pathway) as demonstrated in their in vitro experiments is uncertain. For example, β2-adrenoreceptor activation also causes arteriolar dilation and capillary recruitment in skeletal muscle and thus increases tissue perfusion and (insulin-mediated) glucose uptake (4). Although the current study eloquently provided evidence for β2-adrenoreceptor–mediated glucose uptake in skeletal muscle via the cAMP-mTORC2-GLUT4 pathway at the cellular level, future studies are needed to determine the interactions between cellular and systemic glucose responses following β2-adrenoreceptor activation.
Article
Full-text available
Early hyperglycemia following trauma increases morbidity and mortality. Insulin is widely used to control post-trauma glucose, but this treatment increases the risk of hypoglycemia. We tested a novel method for early post-trauma hyperglycemia control by suppressing hepatic glycogenolysis via β2-adrenoreceptor blockade (ICI 118551, ICI). We have shown that, following severe trauma, obese Zucker rats (OZ), similar with obese patients, exhibit increased acute lung injury (ALI) as compared to lean Zucker rats (LZ). We hypothesize that OZ exhibit a greater increase in early post-trauma glucose as compared to LZ, with the increased post-trauma hyperglycemia suppressed by ICI treatment. Orthopedic trauma was applied to both hindlimbs in LZ and OZ. Fasting plasma glucose was then monitored for 6 hours with or without ICI (0.2mg/kg/hour, iv.) treatment. One day after trauma, plasma IL-6 levels, lung neutrophil numbers, myeloperoxidase (MPO) activity, and wet/dry weight ratios were measured. Trauma induced rapid hepatic glycogenolysis, evidenced by decreased liver glycogen levels, and this was inhibited by ICI treatment. As compared to LZ, OZ exhibited higher post-trauma glucose, IL-6, lung neutrophil infiltration, and MPO activity. Lung wet/dry weight ratios were increased in OZ but not in LZ. ICI treatment reduced the early hyperglycemia, lung neutrophil retention, MPO activity, and wet/dry weight ratio in OZ to levels comparable to those seen in LZ, with no effect on blood pressure or heart rate. These results demonstrate that β2-adrenoreceptor blockade effectively reduces the early post-trauma hyperglycemia, which is associated with decreased lung injury in OZ.
Article
Full-text available
The actin-cytoskeleton-regulating GTPase Rac1 is required for insulin-stimulated GLUT4 translocation in cultured muscle cells. However, involvement of Rac1 and its downstream signaling in glucose transport in insulin sensitive and insulin resistant mature skeletal muscle has not previously been investigated. We hypothesized that Rac1 and its downstream target, p21-activated kinase (PAK), are regulators of insulin-stimulated glucose uptake in mouse and human skeletal muscle, and are dysregulated in insulin resistant states.Muscle specific inducible Rac1 knockout (KO) mice and pharmacological inhibition of Rac1 were used to determine whether Rac1 regulates insulin-stimulated glucose transport in mature skeletal muscle. Furthermore, Rac1 and PAK1 expression and signalling were investigated in muscle of insulin resistant mice and humans.Inhibition and KO of Rac1 decreased insulin-stimulated glucose transport in mouse soleus and EDL muscles ex vivo. Rac1 KO mice showed decreased insulin and glucose tolerance and trended towards higher plasma insulin concentrations following intraperitoneal glucose injection. Rac1 protein expression and PAK(Thr423) phosphorylation were decreased in muscles of high fat fed mice. In humans, insulin-stimulated PAK-activation was decreased in both acute insulin resistant (intralipid infusion) and in chronic insulin resistant states (obesity and diabetes). These findings show that Rac1 is a regulator of insulin-stimulated glucose uptake and a novel candidate involved in skeletal muscle insulin resistance.
Article
Full-text available
In skeletal muscle, the actin cytoskeleton-regulating GTPase, Rac1, is necessary for insulin-dependent GLUT4 translocation. Muscle contraction increases glucose transport and represents an alternative signaling pathway to insulin. Whether Rac1 is activated by muscle contraction and regulates contraction-induced glucose uptake is unknown. Therefore, we studied the effects of in vivo exercise and ex vivo muscle contractions on Rac1 signaling and its regulatory role in glucose uptake in mice and humans. Muscle Rac1-GTP binding was increased after exercise in mice (∼60-100%) and humans (∼40%), and this activation was AMP-activated protein kinase independent. Rac1 inhibition reduced contraction-stimulated glucose uptake in mouse muscle by 55% in soleus and by 20-58% in extensor digitorum longus (EDL; P < 0.01). In agreement, the contraction-stimulated increment in glucose uptake was decreased by 27% (P = 0.1) and 40% (P < 0.05) in soleus and EDL muscles, respectively, of muscle specific inducible Rac1 knockout mice. Furthermore, depolymerization of the actin cytoskeleton decreased contraction-stimulated glucose uptake by 100% and 62% (P < 0.01) in soleus and EDL muscles, respectively. These are the first data to show that Rac1 is activated during muscle contraction in murine and human skeletal muscle and suggest that Rac1 and possibly the actin cytoskeleton are novel regulators of contraction-stimulated glucose uptake.
Article
Full-text available
The activation state of β-adrenergic receptors (β-ARs) in vivo is an important determinant of hemodynamic status, cardiac performance, and metabolic rate. In order to achieve homeostasis in vivo, the cellular signals generated by β-AR activation are integrated with signals from a number of other distinct receptors and signaling pathways. We have utilized genetic knockout models to test directly the role of β1- and/or β2-AR expression on these homeostatic control mechanisms. Despite total absence of β1- and β2-ARs, the predominant cardiovascular β-adrenergic subtypes, basal heart rate, blood pressure, and metabolic rate do not differ from wild type controls. However, stimulation of β-AR function by β-AR agonists or exercise reveals significant impairments in chronotropic range, vascular reactivity, and metabolic rate. Surprisingly, the blunted chronotropic and metabolic response to exercise seen in β1/β2-AR double knockouts fails to impact maximal exercise capacity. Integrating the results from single β1- and β2-AR knockouts as well as the β1-/β2-AR double knock-out suggest that in the mouse, β-AR stimulation of cardiac inotropy and chronotropy is mediated almost exclusively by the β1-AR, whereas vascular relaxation and metabolic rate are controlled by all three β-ARs (β1-, β2-, and β3-AR). Compensatory alterations in cardiac muscarinic receptor density and vascular β3-AR responsiveness are also observed in β1-/β2-AR double knockouts. In addition to its ability to define β-AR subtype-specific functions, this genetic approach is also useful in identifying adaptive alterations that serve to maintain critical physiological setpoints such as heart rate, blood pressure, and metabolic rate when cellular signaling mechanisms are perturbed.
Article
Full-text available
Background: The sympathetic nervous system regulates energy metabolism via β-adrenoreceptors. Therapeutic exploitation of previous β2-adrenegic agonists for metabolic benefits has been hindered by cross stimulation of cardiac β1-adrenoceptor, causing tachycardia. Formoterol is a novel highly β2-selective adrenergic agonist and holds promise as a β2-agonist that could impart selective beneficial metabolic effects. Objective: To investigate the metabolic effects of formoterol on energy and substrate metabolism. Participants: Healthy volunteers. Design: (1) Dose-finding study, step-wise incremental design of weekly administration of 80, 160 and 320 μg daily of formoterol in four subjects and, (2) metabolic study, an open-label metabolic evaluation of 1-week treatment in eight men using a dose determined from (1). Main outcome: Resting energy expenditure (EE), diet-induced thermogenesis (DIT) and fat oxidation (Fox) using indirect calorimetry, heart rate and plasma non-esterified fatty acid (NEFA) levels. Results: In the dose-finding study, all three doses increased resting EE and Fox with the 320 μg dose significantly increasing heart rate. In the metabolic study, the selected 160 μg daily dose increased resting EE by 13 ± 2% (P<0.001) and Fox by 23 ± 4% (P<0.01), but not DIT. Plasma NEFA levels rose by 16 ± 2% (P<0.01). Heart rate did not change significantly. Out of the eight subjects, six reported tremor and palpitation, two lost appetite and one suffered from insomnia. Conclusions: At a dose of 160 μg per day, formoterol increases resting EE and fat utilization without inducing tachycardia. From this first metabolic evaluation in humans, we conclude that formoterol imparts beneficial metabolic changes that may be exploited for therapy of obesity.
Article
Chronic treatment of rats with the ß2-adrenergic agonists clenbuterol and fenoterol over 16–19 d raised energy intake, expenditure, and body weight gain but did not affect fat or energy deposition, and body protein gain was increased by 50 and 18%, respectively. Both drugs increased the protein content and mitochondrial GDP-binding capacity of brown adipose tissue. Clenbuterol did not affect plasma insulin, growth hormone, or triiodothyronine levels, although insulin levels were reduced by fenoterol. Both drugs caused hypertrophy of skeletal muscle (gastrocnemius), and muscle protein synthesis in vivo (fractional rate) was elevated by 34 and 26% in clenbuterol and fenoteroltreated rats, respectively.
Article
Sarcolemma-enriched preparations from muscles rich in slow oxidative red fibres contained specific binding sites for the α1 antagonist, prazosin (e.g. soleus Kd 0.13 nM, Bmax 29 fmol/mg protein). Binding sites for prazosin were almost absent from white muscle. Displacement of prazosin binding from sarcolemma of soleus muscle (phentolamine>phenylephrine>idazoxan>yohimbine) suggested that the receptors were α1. Binding sites for dihydroalprenolol (β antagonist) were also more concentrated on red than white muscle and outnumbered prazosin sites by approx. 10:1. Binding sites for idazoxan (α2 antagonist) were undetectable. Contamination of sarcolemma-enriched preparations by endothelial tissue indicated by the activity of angiotensin converting enzyme did not correlate with prazosin binding. It is concluded that post-synaptic α1 adrenergic receptors are present on the sarcolemma of slow oxidative red fibres of rat skeletal muscle. The presence provides the mechanistic basis for apparent α-adrenergic effects to increase glucose and oxygen uptake in perfused rat hindquarter.
Article
Insulin resistant type 2 diabetes mellitus in the obese elderly has become a worldwide epidemic. While exercise can prevent the onset of diabetes in young subjects its role in older diabetic people is less clear. Exercise stimulates the release of the β(2)-agonist epinephrine more in the young. Although epinephrine and β(2)-agonist drugs cause acute insulin resistance, their chronic effect on insulin sensitivity is unclear. We fed C57BL/6 mice a high fat diet to induce diabetes. These overweight animals became very insulin resistant. Exhaustive treadmill exercise 5 days a week for 8 weeks had no effect on their diabetes, nor did the β(2)-blocking drug ICI 118551. In contrast, exercise combined with the β(2)-agonist salbutamol (albuterol) had a beneficial effect on both glucose tolerance and insulin sensitivity after 4 and 8 weeks of exercise. The effect was durable and persisted 5 weeks after exercise and β(2)-agonist had stopped. To test whether β(2)-agonist alone was effective, the animals that had received β(2)-blockade were then given β(2)-agonist. Their response to a glucose challenge improved but their response to insulin was not significantly altered. The β(2)-agonists are commonly used to treat asthma and asthmatics have an increased incidence of obesity and type 2 diabetes. Although β(2)-agonists cause acute hyperglycemia, chronic treatment improves insulin sensitivity, probably by improving muscle glucose uptake.