Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Natl Cancer Inst ; 107(12): djv274, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26464423

ABSTRACT

BACKGROUND: Cancer cachexia is a debilitating condition that impacts patient morbidity, mortality, and quality of life and for which effective therapies are lacking. The anticachectic activity of the novel HDAC inhibitor AR-42 was investigated in murine models of cancer cachexia. METHODS: The effects of AR-42 on classic features of cachexia were evaluated in the C-26 colon adenocarcinoma and Lewis lung carcinoma (LLC) models. Effects on survival in comparison with approved HDAC inhibitors (vorinostat, romidepsin) were determined. The muscle metabolome and transcriptome (by RNA-seq), as well as serum cytokine profile, were evaluated. Data were analyzed using mixed effects models, analysis of variance, or log-rank tests. All statistical tests were two-sided. RESULTS: In the C-26 model, orally administered AR-42 preserved body weight (23.9±2.6 grams, AR-42-treated; 20.8±1.3 grams, vehicle-treated; P = .005), prolonged survival (P < .001), prevented reductions in muscle and adipose tissue mass, muscle fiber size, and muscle strength and restored intramuscular mRNA expression of the E3 ligases MuRF1 and Atrogin-1 to basal levels (n = 8). This anticachectic effect, confirmed in the LLC model, was not observed after treatment with vorinostat and romidepsin. AR-42 suppressed tumor-induced changes in inflammatory cytokine production and multiple procachexia drivers (IL-6, IL-6Rα, leukemia inhibitory factor, Foxo1, Atrogin-1, MuRF1, adipose triglyceride lipase, uncoupling protein 3, and myocyte enhancer factor 2c). Metabolomic analysis revealed cachexia-associated changes in glycolysis, glycogen synthesis, and protein degradation in muscle, which were restored by AR-42 to a state characteristic of tumor-free mice. CONCLUSIONS: These findings support further investigation of AR-42 as part of a comprehensive therapeutic strategy for cancer cachexia.


Subject(s)
Cachexia/drug therapy , Cytokines/drug effects , Cytokines/metabolism , Histone Deacetylase Inhibitors/pharmacology , Neoplasms, Experimental/complications , Phenylbutyrates/pharmacology , Weight Loss/drug effects , Adenocarcinoma/complications , Adipose Tissue/drug effects , Administration, Oral , Animals , Cachexia/etiology , Cachexia/metabolism , Cachexia/prevention & control , Carcinoma, Lewis Lung/complications , Colonic Neoplasms/complications , Cytokines/biosynthesis , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Interleukin-6/metabolism , Ion Channels/metabolism , Leukemia Inhibitory Factor/metabolism , Lipase/metabolism , MEF2 Transcription Factors/metabolism , Mice , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phenylbutyrates/administration & dosage , Receptors, Interleukin-6/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Survival Analysis , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism , Uncoupling Protein 3
2.
Proc Natl Acad Sci U S A ; 111(12): 4525-9, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24616506

ABSTRACT

MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression and, in cancers, are often packaged within secreted microvesicles. The cachexia syndrome is a debilitating state of cancer that predominantly results from the loss of skeletal muscle mass, which is in part associated with apoptosis. How tumors promote apoptosis in distally located skeletal muscles has not been explored. Using both tumor cell lines and patient samples, we show that tumor-derived microvesicles induce apoptosis of skeletal muscle cells. This proapoptotic activity is mediated by a microRNA cargo, miR-21, which signals through the Toll-like 7 receptor (TLR7) on murine myoblasts to promote cell death. Furthermore, tumor microvesicles and miR-21 require c-Jun N-terminal kinase activity to regulate this apoptotic response. Together, these results describe a unique pathway by which tumor cells promote muscle loss, which might provide a great insight into elucidating the causes and treatment options of cancer cachexia.


Subject(s)
Apoptosis/genetics , Cachexia/pathology , MicroRNAs/physiology , Muscle, Skeletal/pathology , Neoplasms/complications , Organelles/genetics , Toll-Like Receptor 7/physiology , Animals , Cachexia/etiology , Cell Line, Tumor , Humans , Mice , Neoplasms/pathology
3.
J Cachexia Sarcopenia Muscle ; 5(4): 321-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24668658

ABSTRACT

BACKGROUND: Muscle wasting is a profound side effect of advanced cancer. Cancer-induced cachexia decreases patient quality of life and is associated with poor patient survival. Currently, no clinical therapies exist to treat cancer-induced muscle wasting. Although cancers commonly associated with cachexia occur in older individuals, the standard animal models used to elucidate the causes of cachexia rely on juvenile mice. METHODS: In an effort to better model human cancer cachexia, we determined whether cachectic features seen in young mice could be achieved in adult, pre-sarcopenic mice following colon 26 (C-26) tumor cell inoculation. RESULTS: Both young and adult mice developed similar-sized tumors and progressed to cachexia with similar kinetics, as evidenced by losses in body mass, and adipose and skeletal muscle tissues. Proteolytic signaling, including proteasome and autophagy genes, was also increased in muscles from both young and adult tumor-bearing animals. Furthermore, tumor-associated muscle damage and activation of Pax7 progenitor cells was induced in both young and adult mice. CONCLUSIONS: Although cancer cachexia generally occurs in older individuals, these data suggest that the phenotype and underlying mechanisms can be effectively modeled using the currently accepted protocol in juvenile mice.

4.
J Clin Invest ; 123(11): 4821-35, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24084740

ABSTRACT

Cachexia is a debilitating condition characterized by extreme skeletal muscle wasting that contributes significantly to morbidity and mortality. Efforts to elucidate the underlying mechanisms of muscle loss have predominantly focused on events intrinsic to the myofiber. In contrast, less regard has been given to potential contributory factors outside the fiber within the muscle microenvironment. In tumor-bearing mice and patients with pancreatic cancer, we found that cachexia was associated with a type of muscle damage resulting in activation of both satellite and nonsatellite muscle progenitor cells. These muscle progenitors committed to a myogenic program, but were inhibited from completing differentiation by an event linked with persistent expression of the self-renewing factor Pax7. Overexpression of Pax7 was sufficient to induce atrophy in normal muscle, while under tumor conditions, the reduction of Pax7 or exogenous addition of its downstream target, MyoD, reversed wasting by restoring cell differentiation and fusion with injured fibers. Furthermore, Pax7 was induced by serum factors from cachectic mice and patients, in an NF-κB-dependent manner, both in vitro and in vivo. Together, these results suggest that Pax7 responds to NF-κB by impairing the regenerative capacity of myogenic cells in the muscle microenvironment to drive muscle wasting in cancer.


Subject(s)
Cachexia/etiology , Cachexia/metabolism , Muscle, Skeletal/metabolism , NF-kappa B/metabolism , PAX7 Transcription Factor/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cachexia/pathology , Case-Control Studies , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred mdx , Mice, Nude , Mice, Transgenic , Microscopy, Electron, Transmission , Middle Aged , Muscle Development , Muscle, Skeletal/pathology , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , PAX7 Transcription Factor/genetics , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Tumor Microenvironment , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...