30 research outputs found

    Fractional Synthetic Rate and Markers of Protein Turnover are Altered in the Diaphragms of Cachectic Mice

    Get PDF
    Cancer cachexia, a wasting syndrome characterized by rapid skeletal muscle wasting and fat loss, directly accounts for up to 20-40% of cancer-related deaths. All muscles, including respiratory muscles, are susceptible to atrophy because cancer cachexia is a systemic disease. Atrophy of the primary breathing muscle, the diaphragm, can lead to respiratory distress, which is commonly associated with a cachectic phenotype. Indeed, the diaphragm is more susceptible to atrophy in certain conditions, but little is known about the effects of cancer-cachexia on protein turnover in the diaphragm. Therefore, investigations into the alterations in protein turnover could provide insight to the molecular events and provide valuable information in the search for therapeutic targets. PURPOSE: The purpose of this study was to describe changes in diaphragmatic protein synthesis and molecular markers of synthesis and degradation during the progression of cancer cachexia. METHODS: C57BL6/J mice (8 wks old) were implanted with 1X106 Lewis Lung Carcinoma cells (LLC) or Phosphate-Buffered Saline (PBS, control). Tumors developed over a 1-4 wk time course and diaphragms were harvested at each time point (1, 2, 3, or 4 wks). Fractional synthetic rates (FSR) were determined using deuterium incorporation into muscle. Selected markers of protein synthesis and degradation pathways were analyzed by immunoblot analysis. One-Way ANOVA was used for statistical analyses, with significance set at pRESULTS: FSR trended downward over time, but did not reach significance. Similar to FSR, anabolic signaling markers (4EBP-1, ERK1/2, Deptor) did not demonstrate significant differences. p62, an autophagic degradation marker, was significantly less than PBS in 3 wk diaphragms (

    Mitochondrial Dysfunction in Diaphragm Muscle Precedes the Cachectic Phenotype in LLC Tumor-Bearing Mice.

    Get PDF
    The defining feature of cancer cachexia is extensive weight loss and skeletal muscle atrophy. It is clinically important because cachexia reduces patient survival, results in functional impairment, and is estimated to be directly responsible for 20-40% of cancer deaths. Unfortunately, no clinical therapy exists and therefore, it is important to understand the molecular mechanisms responsible for rapid muscle wasting. Compared to limb muscles, the diaphragm is relatively understudied in cancer cachexia, but is likely to be adversely affected because cachexia is a systemic disease. Wasting of the primary inspiratory muscle may result in difficulty breathing and inability to adjust minute ventilation in response to a respiratory challenge. Based on emerging evidence, it is clear that oxidative stress is present in cachexia-induced wasting of the diaphragm; PURPOSE: we developed the hypothesis that mitochondrial dysfunction in the diaphragm precedes cachexia. METHODS: 1X106 Lewis Lung Carcinoma cells (LLC) or Phosphate-Buffered Saline (PBS, control) were implanted to the hind-flank of C57BL6/J mice at 8 wks of age. Tumors were allowed to develop for 1, 2, 3, or 4 wks. At designated time points diaphragms were collected and mitochondrial function was assessed by respiration and ROS production. RESULTS: Cancer cachexia was evident only at the 4 wk time point demonstrated by decrease in body mass and muscle atrophy in several limb muscles. Mitochondrial respiration, assessed by respiratory control ratio (state3/state 4 respiration), was significantly lower at 1 wk (pCONCLUSIONS:The molecular events that lead to muscle atrophy in cancer cachexia are unknown. We demonstrate that two hallmarks of mitochondrial dysfunction, altered respiration and ROS production, occur in the diaphragm well before the cancer cachexia phenotype is evident in the LLC model. These data suggest that the mitochondria are likely a suitable target to treat or prevent cancer cachexia-induced muscle wasting in the diaphragm

    Mitochondrial Dysfunction is Evident in Lewis Lung Carcinoma-Induced Muscle Wasting

    Get PDF
    Cancer cachexia is a paraneoplastic syndrome associated with adverse prognosis and shortened survival. The defining feature of cachexia is extensive muscle atrophy leading to progressive functional impairments. The molecular mechanisms responsible for the rapid muscle wasting are not fully elucidated. Based on emerging evidence, we developed the hypothesis cachectic muscle wasting is caused by mitochondrial dysfunction increasing reactive oxygen species production leading to global oxidative stress. To test this hypothesis we utilized the well-established Lewis-Lung Carcinoma (LLC) model of cancer cachexia. The time-course study consisted of one, two, three and four week LLC tumor bearing mice and age-matched four week saline (PBS) control (Ctrl) mice. Tumors were implanted into the hind flank at 1X106 cells in 100 µL PBS. The plantaris was weighed for wet mass then teased into small fiber bundles and permeabilized for the quantification of mitochondrial function. Mitochondrial dysfunction was classified by a decrease in the respiratory control ratio (RCR), which is the ratio of state 3 (maximal ADP stimulated respiration) to state 4 (oligomycin-induced leak respiration). Muscle mass progressively declined over the time-course, reaching significance at 4 weeks (Ctrl vs 4-week, p\u3c0.05). Mitochondrial function was not different among groups, however individual a priori comparison between groups revealed that 4wk cancer animals exhibited marked mitochondrial dysfunction compared to all other groups (p\u3c0.05). These data demonstrate that late stage cancer-induced muscle wasting is associated with significant mitochondrial dysfunction

    Disuse Atrophy Occurs Without a Change in Mitochondrial Respiratory Control Ratio During Hindlimb Unloading in Mice

    Get PDF
    Skeletal muscle atrophy commonly occurs during prolonged periods of inactivity, however, the precise mechanisms that cause muscle atrophy have yet to be determined. Specifically, a controversy exists on whether mitochondrial dysfunction is a cause or consequence of disuse muscle atrophy. PURPOSE: The purpose of this study was to determine if a change in the respiratory control ratio, which is a ratio of maximal O2 respiration to leak respiration, could be detected prior to muscle atrophy in a time-course study in mice. METHODS: Disuse atrophy was induced using hindlimb unloading (HU) in adult, C57BL/6J male mice for 0 (control), 1, 2, 3, or 7 days (n=~6-8/group). Following completion, gastrocnemius and soleus muscles were weighed and assessed for mitochondrial function in permeabilized muscle fibers. Here, we define mitochondrial function as the respiratory control ratio (RCR) determined by maximal ADP stimulated respiration (State 3) divided by leak or ATP synthase inhibited (state 4) respiration. A one-way ANOVA was used to determine differences between means. When significant F ratios were found, a Tukey post-hoc was used to compare differences between means. Values presented are mean ± standard error RESULTS: In both the gastrocnemius and soleus, muscle mass was not significantly different from control at day 1, but was significantly lower at 2, 3, and 7-day timepoints. In contrast, there was no significant difference in RCR in gastrocnemius (control 3.11±0.20, 1 day 2.96±0.60, 2 day 3.07±0.31, 3 day 3.08±0.25, 7 day 3.41± .29) or soleus (control 2.33±0.33, 1 day 2.77±0.33, 2 day 3.03±0.51, 3 day 2.93±0.30, 7 day 2.78±0.48). CONCLUSION: It is well established that HU causes rapid muscle atrophy. These data support mitochondrial RCR does decrease before muscle atrophy in either gastrocnemius or soleus muscle, and therefore may not be a primary cause of HU-induced muscle atrophy in mice

    Males, but Not Females, Demonstrate Mitochondrial Dysfunction in the C26 Model of Cancer Cachexia

    Get PDF
    Cancer cachexia is characterized by progressive muscle wasting that can lead to symptoms such as anemia, severe weight loss, and fatigue. These symptoms can lead to limitations in activities of daily living and can cause resistance to chemotherapy treatments in cancer patients. There are no current treatments available to treat cancer cachexia and a critical need remains to identify mechanisms of cancer cachexia. Recently, our group identified mitochondrial disfunction precedes muscle atrophy in males but not females in a model of lung cancer induced atrophy. However, it is unknown whether this finding is replicated when studying a different type of cancer. PURPOSE: This study set out to determine if mitochondrial respiration is impaired in the plantaris muscle in a well-established colon cancer model of cachexia. METHODS: The time-course study consisted of male and female mice in four groups per sex: An age-matched control (PBS), and three groups implanted with C26 tumors. Tumor growth for 10-15 days, 20 days, and 25 days. Tumors were implanted bilaterally into the hind flank for a total of 1X106 cells PBS (one-half per each hindflank). The plantaris was weighed for wet mass then teased into small fiber bundles and permeabilized for the quantification of mitochondrial function. Mitochondrial dysfunction was classified by a decrease in the respiratory control ratio (RCR), which is the ratio of state 3 (maximal ADP stimulated respiration) to state 4 (oligomycin-induced leak respiration). Male and Female data were analyzed separately using a one-way ANOVA. RESULTS: The tumor burden increased as the number of days increased. Male RCR showed a mean difference in RCR at the early timepoint (10-15 day, p=0.058) and demonstrated significantly lower RCR at the 20 day timepoint compared to PBS control (20d= 1.170± 0.094, PBS= 2.41 ± 0.13, p=0.031). Interestingly, RCR was not significantly different between male PBS and 25 days (1.864± 0.21, p=0.084). RCR in the plantaris from females was not different among any of the groups (p=0.401). CONCLUSION: Along with our previously published data in a lung cancer model, these data indicate that the mechanisms of muscle atrophy are sex dependent. Specifically, mitochondrial dysfunction appears to play an important role in cancer-induced atrophy in male, but not female, mice

    Mitochondrial function and protein turnover in the diaphragm are altered in llc tumor model of cancer cachexia

    Get PDF
    It is established that cancer cachexia causes limb muscle atrophy and is strongly associated with morbidity and mortality; less is known about how the development of cachexia impacts the diaphragm. The purpose of this study was to investigate cellular signaling mechanisms related to mitochondrial function, reactive oxygen species (ROS) production, and protein synthesis during the development of cancer cachexia. C57BL/J6 mice developed Lewis Lung Carcinoma for either 0 weeks (Control), 1 week, 2 weeks, 3 weeks, or 4 weeks. At designated time points, diaphragms were harvested and analyzed. Mitochondrial respiratory control ratio was ~50% lower in experimental groups, which was significant by 2 weeks of cancer development, with no difference in mitochondrial content markers COXIV or VDAC. Compared to the controls, ROS was 4-fold elevated in 2-week animals but then was not different at later time points. Only one antioxidant protein, GPX3, was altered by cancer development (~70% lower in experimental groups). Protein synthesis, measured by a fractional synthesis rate, appeared to become progressively lower with the cancer duration, but the mean difference was not significant. The development and progression of cancer cachexia induces marked alterations to mitochondrial function and ROS production in the diaphragm and may contribute to increased cachexia-associated morbidity and mortality

    Mitochondrial degeneration precedes the development of muscle atrophy in progression of cancer cachexia in tumour-bearing mice

    Get PDF
    Background: Cancer cachexia is largely irreversible, at least via nutritional means, and responsible for 20–40% of cancer-related deaths. Therefore, preventive measures are of primary importance; however, little is known about muscle perturbations prior to onset of cachexia. Cancer cachexia is associated with mitochondrial degeneration; yet, it remains to be determined if mitochondrial degeneration precedes muscle wasting in cancer cachexia. Therefore, our purpose was to determine if mitochondrial degeneration precedes cancer-induced muscle wasting in tumour-bearing mice. Methods: First, weight-stable (MinStable) and cachectic (MinCC) ApcMin/+ mice were compared with C57Bl6/J controls for mRNA contents of mitochondrial quality regulators in quadriceps muscle. Next, Lewis lung carcinoma (LLC) cells or PBS (control) were injected into the hind flank of C57Bl6/J mice at 8 week age, and tumour allowed to develop for 1, 2, 3, or 4 weeks to examine time course of cachectic development. Succinate dehydrogenase stain was used to measure oxidative phenotype in tibialis anterior muscle. Mitochondrial quality and function were assessed using the reporter MitoTimer by transfection to flexor digitorum brevis and mitochondrial function/ROS emission in permeabilized adult myofibres from plantaris. RT-qPCR and immunoblot measured the expression of mitochondrial quality control and antioxidant proteins. Data were analysed by one-way ANOVA with Student–Newman–Kuels post hoc test. Results: MinStable mice displayed ~50% lower Pgc-1α, Pparα, and Mfn2 compared with C57Bl6/J controls, whereas MinCC exhibited 10-fold greater Bnip3 content compared with C57Bl6/J controls. In LLC, cachectic muscle loss was evident only at 4 weeks post-tumour implantation. Oxidative capacity and mitochondrial content decreased by ~40% 4 weeks post-tumour implantation. Mitochondrial function decreased by ~25% by 3 weeks after tumour implantation. Mitochondrial degeneration was evident by 2 week LLC compared with PBS control, indicated by MitoTimer red/green ratio and number of pure red puncta. Mitochondrial ROS production was elevated by ~50 to ~100% when compared with PBS at 1–3 weeks post-tumour implantation. Mitochondrial quality control was dysregulated throughout the progression of cancer cachexia in tumour-bearing mice. In contrast, antioxidant proteins were not altered in cachectic muscle wasting. Conclusions: Functional mitochondrial degeneration is evident in LLC tumour-bearing mice prior to muscle atrophy. Contents of mitochondrial quality regulators across ApcMin/+ and LLC mice suggest impaired mitochondrial quality control as a commonality among pre-clinical models of cancer cachexia. Our data provide novel evidence for impaired mitochondrial health prior to cachectic muscle loss and provide a potential therapeutic target to prevent cancer cachexia

    Female mice may have exacerbated catabolic signalling response compared to male mice during development and progression of disuse atrophy

    Get PDF
    Background: Muscle atrophy is a common pathology associated with disuse, such as prolonged bed rest or spaceflight, and is associated with detrimental health outcomes. There is emerging evidence that disuse atrophy may differentially affect males and females. Cellular mechanisms contributing to the development and progression of disuse remain elusive, particularly protein turnover cascades. The purpose of this study was to investigate the initial development and progression of disuse muscle atrophy in male and female mice using the well-established model of hindlimb unloading (HU). Methods: One hundred C57BL/6J mice (50 male and 50 female) were hindlimb suspended for 0 (control), 24, 48, 72, or 168 h to induce disuse atrophy (10 animals per group). At designated time points, animals were euthanized, and tissues (extensor digitorum longus, gastrocnemius, and soleus for mRNA analysis, gastrocnemius and extensor digitorum longus for protein synthesis rates, and tibialis anterior for histology) were collected for analysis of protein turnover mechanisms (protein anabolism and catabolism). Results: Both males and females lost ~30% of tibialis anterior cross-sectional area after 168 h of disuse. Males had no statistical difference in MHCIIB fibre area, whereas unloaded females had ~33% lower MHCIIB cross-sectional area by 168 h of unloading. Both males and females had lower fractional protein synthesis rates (FSRs) within 24-48 h of HU, and females appeared to have a greater reduction compared with males within 24 h of HU (~23% lower FSRs in males vs. 40% lower FSRs in females). Males and females exhibited differential patterns and responses in multiple markers of protein anabolism, catabolism, and myogenic capacity during the development and progression of disuse atrophy. Specifically, females had greater mRNA inductions of catabolic factors Ubc and Gadd45a (~4-fold greater content in females compared with ~2-fold greater content in males) and greater inductions of anabolic inhibitors Redd1 and Deptor with disuse across multiple muscle tissues exhibiting different fibre phenotypes. Conclusions: These results suggest that the aetiology of disuse muscle atrophy is more complicated and nuanced than previously thought, with different responses based on muscle phenotypes and between males and females, with females having greater inductions of atrophic markers early in the development of disuse atrophy

    Exercise Counteracts the Deleterious Effects of Cancer Cachexia

    No full text
    Cancer cachexia (CC) is a multifactorial syndrome characterised by unintentional loss of body weight and muscle mass in patients with cancer. The major hallmarks associated with CC development and progression include imbalanced protein turnover, inflammatory signalling, mitochondrial dysfunction and satellite cell dysregulation. So far, there is no effective treatment to counteract muscle wasting in patients with CC. Exercise training has been proposed as a potential therapeutic approach for CC. This review provides an overview of the effects of exercise training in CC-related mechanisms as well as how factors such as cancer comorbidities, exercise modality and biological sex can influence exercise effectiveness in CC. Evidence in mice and humans suggests exercise training combats all of the hallmarks of CC. Several exercise modalities induce beneficial adaptations in patients/animals with CC, but concurrent resistance and endurance training is considered the optimal type of exercise. In the case of cancer patients presenting comorbidities, exercise training should be performed only under specific guidelines and precautions to avoid adverse effects. Observational comparison of studies in CC using different biological sex shows exercise-induced adaptations are similar between male and female patients/animals with cancer, but further studies are needed to confirm this
    corecore