Network


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

Hotspot


Dive into the research topics where Michael P. Wiggs is active.

Publication


Featured researches published by Michael P. Wiggs.


American Journal of Physiology-endocrinology and Metabolism | 2012

Mitochondrial signaling contributes to disuse muscle atrophy.

Scott K. Powers; Michael P. Wiggs; José Alberto Duarte; A. Murat Zergeroglu; Haydar A. Demirel

It is well established that long durations of bed rest, limb immobilization, or reduced activity in respiratory muscles during mechanical ventilation results in skeletal muscle atrophy in humans and other animals. The idea that mitochondrial damage/dysfunction contributes to disuse muscle atrophy originated over 40 years ago. These early studies were largely descriptive and did not provide unequivocal evidence that mitochondria play a primary role in disuse muscle atrophy. However, recent experiments have provided direct evidence connecting mitochondrial dysfunction to muscle atrophy. Numerous studies have described changes in mitochondria shape, number, and function in skeletal muscles exposed to prolonged periods of inactivity. Furthermore, recent evidence indicates that increased mitochondrial ROS production plays a key signaling role in both immobilization-induced limb muscle atrophy and diaphragmatic atrophy occurring during prolonged mechanical ventilation. Moreover, new evidence reveals that, during denervation-induced muscle atrophy, increased mitochondrial fragmentation due to fission is a required signaling event that activates the AMPK-FoxO3 signaling axis, which induces the expression of atrophy genes, protein breakdown, and ultimately muscle atrophy. Collectively, these findings highlight the importance of future research to better understand the mitochondrial signaling mechanisms that contribute to disuse muscle atrophy and to develop novel therapeutic interventions for prevention of inactivity-induced skeletal muscle atrophy.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2013

Ventilator-induced diaphragm dysfunction: cause and effect

Scott K. Powers; Michael P. Wiggs; Kurt J. Sollanek; Ashley J. Smuder

Mechanical ventilation (MV) is used clinically to maintain gas exchange in patients that require assistance in maintaining adequate alveolar ventilation. Common indications for MV include respiratory failure, heart failure, drug overdose, and surgery. Although MV can be a life-saving intervention for patients suffering from respiratory failure, prolonged MV can promote diaphragmatic atrophy and contractile dysfunction, which is referred to as ventilator-induced diaphragm dysfunction (VIDD). This is significant because VIDD is thought to contribute to problems in weaning patients from the ventilator. Extended time on the ventilator increases health care costs and greatly increases patient morbidity and mortality. Research reveals that only 18-24 h of MV is sufficient to develop VIDD in both laboratory animals and humans. Studies using animal models reveal that MV-induced diaphragmatic atrophy occurs due to increased diaphragmatic protein breakdown and decreased protein synthesis. Recent investigations have identified calpain, caspase-3, autophagy, and the ubiquitin-proteasome system as key proteases that participate in MV-induced diaphragmatic proteolysis. The challenge for the future is to define the MV-induced signaling pathways that promote the loss of diaphragm protein and depress diaphragm contractility. Indeed, forthcoming studies that delineate the signaling mechanisms responsible for VIDD will provide the knowledge necessary for the development of a pharmacological approach that can prevent VIDD and reduce the incidence of weaning problems.


Free Radical Research | 2014

Exercise-induced improvements in myocardial antioxidant capacity: the antioxidant players and cardioprotection

Scott K. Powers; Kurt J. Sollanek; Michael P. Wiggs; Haydar A. Demirel; Ashley J. Smuder

Abstract Endurance exercise training is known to promote beneficial adaptations to numerous tissues including the heart. Indeed, endurance exercise training results in a cardioprotective phenotype that resists injury during an ischemia–reperfusion (IR) insult. Because IR-induced cardiac injury is due, in part, to increased production of radicals and other reactive oxygen species, many studies have explored the impact of exercise training on myocardial antioxidant capacity. Unfortunately, the literature describing the effects of exercise on the cardiac antioxidant capacity is widely inconsistent. Nonetheless, a growing body of evidence indicates that regular bouts of endurance exercise promote an increase in the expression of both superoxide dismutase 1 and 2 in cardiac mitochondria. Moreover, emerging evidence suggests that exercise also increases accessory antioxidant enzymes in the heart. Importantly, robust evidence indicates that as few as five consecutive days of endurance exercise training results in a cardiac phenotype that resists IR-induced arrhythmias, myocardial stunning, and infarction. Further, mechanistic studies indicate that exercise-induced increases in mitochondrial superoxide dismutase 2 play a key role in this adaptation. Future studies are required to provide a complete picture regarding the cellular adaptations that are responsible for exercise-induced cardioprotection.


The FASEB Journal | 2013

Abnormal protein turnover and anabolic resistance to exercise in sarcopenic obesity

Mats I. Nilsson; Justin P. Dobson; Nicholas P. Greene; Michael P. Wiggs; Kevin L. Shimkus; Elyse V. Wudeck; Amanda R. Davis; Marissa Laureano; James D. Fluckey

Obesity may impair protein synthesis rates and cause anabolic resistance to growth factors, hormones, and exercise, ultimately affecting skeletal muscle mass and function. To better understand muscle wasting and anabolic resistance with obesity, we assessed protein 24‐h fractional synthesis rates (24‐h FSRs) in selected hind‐limb muscles of sedentary and resistance‐exercised lean and obese Zucker rats. Despite atrophied hind‐limb muscles (–28% vs. lean rats), 24‐h FSRs of mixed proteins were significantly higher in quadriceps (+18%) and red or white gastrocnemius (+22 or +38%, respectively) of obese animals when compared to lean littermates. Basal synthesis rates of myofibrillar (+8%) and mitochondrial proteins (–1%) in quadriceps were not different between phenotypes, while manufacture of cytosolic proteins (+12%) was moderately elevated in obese cohorts. Western blot analyses revealed a robust activation of p70S6k (+178%) and a lower expression of the endogenous mTOR inhibitor DEPTOR (–28%) in obese rats, collectively suggesting that there is an obesity‐induced increase in net protein turnover favoring degradation. Lastly, the protein synthetic response to exercise of mixed (–7%), myofibrillar (+6%), and cytosolic (+7%) quadriceps subfractions was blunted compared to the lean phenotype (+34, +40, and +17%, respectively), indicating a muscle‐ and subfraction‐specific desensitization to the anabolic stimulus of exercise in obese animals.—Nilsson, M. I., Dobson, J. P., Greene, N. P., Wiggs, M. P., Shimkus, K. L., Wudeck, E. V., Davis, A. R., Laureano, M. L., Fluckey, J. D., Abnormal protein turnover and anabolic resistance to exercise in sarcopenic obesity. FASEB J. 27, 3905–3916 (2013). www.fasebj.org


The Journal of Physiology | 2015

Increased mitochondrial emission of reactive oxygen species and calpain activation are required for doxorubicin‐induced cardiac and skeletal muscle myopathy

Kisuk Min; Oh-Sung Kwon; Ashley J. Smuder; Michael P. Wiggs; Kurt J. Sollanek; Demetra D. Christou; Jeung-Ki Yoo; Moon-Hyon Hwang; Hazel H. Szeto; Andreas N. Kavazis; Scott K. Powers

Although doxorubicin is a highly effective anti‐tumour agent, the administration of this drug is associated with significant side effects, including contractile dysfunction and myopathy of both cardiac and skeletal muscles. The mechanism(s) responsible for doxorubicin‐induced contractile dysfunction and myopathy in cardiac and skeletal muscles remains unclear. In the present study, we report that increased mitochondrial oxidant production and calpain activation are major contributors to the development of doxorubicin‐induced myopathy. Moreover, treatment with a mitochondrial‐targeted peptide protects against doxorubicin‐induced mitochondrial dysfunction and myopathy in both heart and skeletal muscles. These experiments provide insight into the mechanisms responsible for DOX‐induced contractile dysfunction and myopathy in cardiac and skeletal muscles. Importantly, our results may provide the basis for developing therapeutic approaches to prevent doxorubicin‐induced cardiac and skeletal muscle myopathy.


Metabolism-clinical and Experimental | 2012

Acute resistance exercise augments integrative myofibrillar protein synthesis

Heath G. Gasier; James D. Fluckey; Stephen F. Previs; Michael P. Wiggs; Steven E. Riechman

The purpose of this study was to determine whether an acute bout of high-intensity resistance exercise (RE) would augment integrative mixed muscle and myofibrillar protein fractional synthesis rates (FSRs) when total energy and macronutrient intake was controlled. Twelve healthy young men were studied over 24 hours and performed an acute bout of exhaustive (5 sets until volitional failure of their 85% 1-repetition maximum) unilateral leg press and knee extension exercise, such that one leg was exercised (EX) and the other served as a control (CON). (2)H(2)O (70%) was provided to measure mixed muscle and myofibrillar FSR, and muscle biopsies (vastus lateralis) were collected from the EX and CON legs 16 hours following the RE session. (2)H-labeling of body water over the course of the experiment was 0.32 ± 0.01 mole percent excess. Interestingly, integrative mixed muscle FSR (percent per hour) was similar between the CON (0.76% ± 0.08%) and EX (0.69% ± 0.06%) legs. In contrast, upon determination of myofibrillar FSR, there was an RE effect (EX, 0.94% ± 0.16% vs CON, 0.75% ± 0.08%; P < .05). High-intensity RE without prior training impacts integrative myofibrillar 24-hour FSR, perhaps without altering total responses.


American Journal of Physiology-endocrinology and Metabolism | 2010

Insulin resistance syndrome blunts the mitochondrial anabolic response following resistance exercise

Mats I. Nilsson; Nicholas P. Greene; Justin P. Dobson; Michael P. Wiggs; Heath G. Gasier; Brandon R. Macias; Kevin L. Shimkus; James D. Fluckey

Metabolic risk factors associated with insulin resistance syndrome may attenuate augmentations in skeletal muscle protein anabolism following contractile activity. The purpose of this study was to investigate whether or not the anabolic response, as defined by an increase in cumulative fractional protein synthesis rates (24-h FSR) following resistance exercise (RE), is blunted in skeletal muscle of a well-established rodent model of insulin resistance syndrome. Four-month-old lean (Fa/?) and obese (fa/fa) Zucker rats engaged in four lower body RE sessions over 8 days, with the last bout occurring 16 h prior to muscle harvest. A priming dose of deuterium oxide ((2)H(2)O) and (2)H(2)O-enriched drinking water were administered 24 h prior to euthanization for assessment of cumulative FSR. Fractional synthesis rates of mixed (-5%), mitochondrial (-1%), and cytosolic (+15%), but not myofibrillar, proteins (-16%, P = 0.012) were normal or elevated in gastrocnemius muscle of unexercised obese rats. No statistical differences were found in the anabolic response of cytosolic and myofibrillar subfractions between phenotypes, but obese rats were not able to augment 24-h FSR of mitochondria to the same extent as lean rats following RE (+14% vs. +28%, respectively). We conclude that the mature obese Zucker rat exhibits a mild, myofibrillar-specific suppression in basal FSR and a blunted mitochondrial response to contractile activity in mixed gastrocnemius muscle. These findings underscore the importance of assessing synthesis rates of specific myocellular subfractions to fully elucidate perturbations in basal protein turnover rates and differential adaptations to exercise stimuli in metabolic disease.


The FASEB Journal | 2014

Inhibition of Janus kinase signaling during controlled mechanical ventilation prevents ventilation-induced diaphragm dysfunction

Ira J. Smith; Guillermo L. Godinez; Baljit Singh; Kelly McCaughey; Raniel R. Alcantara; Tarikere L. Gururaja; Melissa S. Ho; Henry N. Nguyen; Annabelle M. Friera; Kathy White; John R. McLaughlin; Derek Hansen; Jason Romero; Kristen A. Baltgalvis; Mark D. Claypool; Wei Li; Wayne Lang; George C. Yam; Marina Gelman; Rongxian Ding; Stephanie Yung; Daniel P. Creger; Yan Chen; Rajinder Singh; Ashley J. Smuder; Michael P. Wiggs; Oh-Sung Kwon; Kurt J. Sollanek; Scott K. Powers; Esteban Masuda

Controlled mechanical ventilation (CMV) is associated with the development of diaphragm atrophy and contractile dysfunction, and respiratory muscle weakness is thought to contribute significantly to delayed weaning of patients. Therefore, therapeutic strategies for preventing these processes may have clinical benefit. The aim of the current study was to investigate the role of the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in CMV‐mediated diaphragm wasting and weakness in rats. CMV‐induced diaphragm atrophy and contractile dysfunction coincided with marked increases in STAT3 phosphorylation on both tyrosine 705 (Tyr705) and serine 727 (Ser727). STAT3 activation was accompanied by its translocation into mitochondria within diaphragm muscle and mitochondrial dysfunction. Inhibition of JAK signaling during CMV prevented phosphorylation of both target sites on STAT3, eliminated the accumulation of phosphorylated STAT3 within the mitochondria, and reversed the pathologic alterations in mitochondrial function, reduced oxidative stress in the diaphragm, and maintained normal diaphragm contractility. In addition, JAK inhibition during CMV blunted the activation of key proteolytic pathways in the diaphragm, as well as diaphragm atrophy. These findings implicate JAK/STAT3 signaling in the development of diaphragm muscle atrophy and dysfunction during CMV and suggest that the delayed extubation times associated with CMV can be prevented by inhibition of Janus kinase signaling.—Smith, I. J., Godinez, G. L., Singh, B. K., McCaughey, K. M., Alcantara, R. R., Gururaja, T., Ho, M. S., Nguyen, H. N., Friera, A. M., White, K. A., McLaughlin, J. R., Hansen, D., Romero, J. M., Baltgalvis, K. A., Claypool, M. D., Li, W., Lang, W., Yam, G. C., Gelman, M. S., Ding, R., Yung, S. L., Creger, D. P., Chen, Y., Singh, R., Smuder, A. J., Wiggs, M. P., Kwon, O.‐S., Sollanek, K. J., Powers, S. K., Masuda, E. S., Taylor, V. C., Payan, D. G., Kinoshita, T., Kinsella, T. M. Inhibition of Janus kinase signaling during controlled mechanical ventilation prevents ventilation‐induced diaphragm dysfunction. FASEB J. 28, 2790–2803 (2014). www.fasebj.org


Acta Physiologica | 2011

Cumulative responses of muscle protein synthesis are augmented with chronic resistance exercise training.

Heath G. Gasier; Steven E. Riechman; Michael P. Wiggs; A. Buentello; Stephen F. Previs; James D. Fluckey

Aim:  The purpose of this study was to determine the anabolic response of a single bout of high intensity resistance exercise (RE) following 5 weeks of RE training.


Frontiers in Physiology | 2015

Can endurance exercise preconditioning prevention disuse muscle atrophy

Michael P. Wiggs

Emerging evidence suggests that exercise training can provide a level of protection against disuse muscle atrophy. Endurance exercise training imposes oxidative, metabolic, and heat stress on skeletal muscle which activates a variety of cellular signaling pathways that ultimately leads to the increased expression of proteins that have been demonstrated to protect muscle from inactivity –induced atrophy. This review will highlight the effect of exercise-induced oxidative stress on endogenous enzymatic antioxidant capacity (i.e., superoxide dismutase, glutathione peroxidase, and catalase), the role of oxidative and metabolic stress on PGC1-α, and finally highlight the effect heat stress and HSP70 induction. Finally, this review will discuss the supporting scientific evidence that these proteins can attenuate muscle atrophy through exercise preconditioning.

Collaboration


Dive into the Michael P. Wiggs's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aaron B. Morton

University of West Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge