Cachexia is characterized by progressive skeletal muscle and body weight loss and affects up to 80% of cancer patients. Since this loss of muscle mass contributes to weakness, reduced tolerance to conventional treatments, and increased mortality, understanding the mechanisms that drive muscle wasting is critical to the development of treatments to improve quality of life and enhance survival of cancer patients. Published and unpublished data from our lab have identified the Forkhead Box (Fox) Class O protein, FoxO1, as a critical regulator of muscle wasting that is associated with cachexia in cancer patients. We identified the transcriptional repressor, FoxP1, as a target gene upregulated by FoxO1 in response to tumor burden (TBu) that is increased at time points which precede and parallel muscle wasting and is also increased in muscle of cachectic cancer patients. In preliminary experiments we found that FoxP1 a) may act as a potent repressor of key transcription factors and genes involved in muscle contraction and the maintenance of muscle integrity, b) is sufficient to induce muscle fiber atrophy and c) is required for muscle fiber atrophy induced by TBu, thus implicating FoxP1 in the cachectic phenotype.
Aims 1 and 2 will therefore test our underlying hypothesis that FoxP1 is sufficient to induce deterioration of muscle ultrastructure, activation of catabolic pathways, and muscle atrophy and weakness and that its upregulation in response to TBu is required for these tumor-induced muscle pathologies.
Specific Aim 1 : We will establish the role and investigate the molecular mechanisms by which FoxP1 is sufficient to induce muscle wasting.
Specific Aim 2 : To establish the role and investigate the mechanisms by which a FoxO1-FoxP1-MEF2c axis drives cancer-induced muscle wasting. We will test these hypotheses in a conventional pre-clinical mouse model of cachexia as well as in a novel mouse model of cancer cachexia in which primary resected tumors from pancreatic ductal adenocarcinoma (PDAC) patients are attached to the pancreas of immunocompromised mice to create mouse avatars.
Specific Aim 3 : To identify changes in the skeletal muscle transcriptome and proteome of weight losing PDAC patients and their mouse avatars during the progression of cancer cachexia.
Our third aim takes advantage of our wide access to muscle and tumor tissue from PDAC patients. We thus propose an unbiased approach to identify the transcriptome (via RNA-seq) and proteome (via iTRAQ) in muscle of PDAC patients as well as their mouse avatars during the progression of cachexia as a surrogate for longitudinal studies in humans. We propose to complement these findings with muscle histology and ultrastructure. We anticipate that our findings from Aims 1 and 2 will elucidate key mechanisms of wasting and weakness in response to TBu, while data generated in Aim 3 will generate a plethora of new knowledge to guide future pre- clinical and clinical studies aimed at preventing or retarding cancer-induced cachexia in the human condition.

Public Health Relevance

Skeletal muscle wasting and weakness are associated with several cancers. Given that skeletal muscle makes up about 40% of body mass, cancer associated muscle wasting typically presents as whole body wasting and is referred to as cachexia. Cachexia is particularly prevalent in pancreatic cancer, affecting up to 80% of patients. It has a significant impact on physical function and quality of life, but also impacts patient?s ability to withstand the dose and frequency of aggressive conventional treatments for the disease and thus impacts survival. We have identified a potent repressor protein called Forkhead boxP1 (FoxP1) which is increased in muscle of pancreatic cancer patients and will study the role of this protein in the wasting process. Identifying novel therapeutic targets to counter cancer-induced muscle atrophy and weakness could represent a major breakthrough for this debilitating condition.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR060209-08
Application #
9769507
Study Section
Skeletal Muscle and Exercise Physiology Study Section (SMEP)
Program Officer
Boyce, Amanda T
Project Start
2011-08-01
Project End
2022-08-31
Budget Start
2019-09-01
Budget End
2020-08-31
Support Year
8
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of Florida
Department
Other Health Professions
Type
Schools of Public Health
DUNS #
969663814
City
Gainesville
State
FL
Country
United States
Zip Code
32611
Kandarian, Susan C; Nosacka, Rachel L; Delitto, Andrea E et al. (2018) Tumour-derived leukaemia inhibitory factor is a major driver of cancer cachexia and morbidity in C26 tumour-bearing mice. J Cachexia Sarcopenia Muscle 9:1109-1120
Murphy, Kate T; Hossain, Mohammed I; Swiderski, Kristy et al. (2018) Mas receptor activation slows tumor growth and attenuates muscle wasting in cancer. Cancer Res :
Delitto, Daniel; Judge, Sarah M; Delitto, Andrea E et al. (2017) Human pancreatic cancer xenografts recapitulate key aspects of cancer cachexia. Oncotarget 8:1177-1189
Ahn, Bumsoo; Beharry, Adam W; Frye, Gregory S et al. (2015) NAD(P)H oxidase subunit p47phox is elevated, and p47phox knockout prevents diaphragm contractile dysfunction in heart failure. Am J Physiol Lung Cell Mol Physiol 309:L497-505
Ryder, Daniel J; Judge, Sarah M; Beharry, Adam W et al. (2015) Identification of the Acetylation and Ubiquitin-Modified Proteome during the Progression of Skeletal Muscle Atrophy. PLoS One 10:e0136247
Beharry, Adam W; Judge, Andrew R (2015) Differential expression of HDAC and HAT genes in atrophying skeletal muscle. Muscle Nerve 52:1098-101
Reid, Michael B; Judge, Andrew R; Bodine, Sue C (2014) Rebuttal from Michael B. Reid, Andrew R. Judge and Sue C. Bodine. J Physiol 592:5351
Reid, Michael B; Judge, Andrew R; Bodine, Sue C (2014) CrossTalk opposing view: The dominant mechanism causing disuse muscle atrophy is proteolysis. J Physiol 592:5345-7
Beharry, Adam W; Sandesara, Pooja B; Roberts, Brandon M et al. (2014) HDAC1 activates FoxO and is both sufficient and required for skeletal muscle atrophy. J Cell Sci 127:1441-53
Judge, Sarah M; Wu, Chia-Ling; Beharry, Adam W et al. (2014) Genome-wide identification of FoxO-dependent gene networks in skeletal muscle during C26 cancer cachexia. BMC Cancer 14:997

Showing the most recent 10 out of 16 publications