Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Article in English | MEDLINE | ID: mdl-38632714

ABSTRACT

BACKGROUND: Patients with pancreatic ductal adenocarcinoma (PDAC) often suffer from cachexia, a wasting syndrome that significantly reduces both quality of life and survival. Although advanced cachexia is associated with inflammatory signalling and elevated muscle catabolism, the early events driving wasting are poorly defined. During periods of nutritional scarcity, the body relies on hepatic ketogenesis to generate ketone bodies, and lipid metabolism via ketogenesis is thought to protect muscle from catabolizing during nutritional scarcity. METHODS: We developed an orthotopic mouse model of early PDAC cachexia in 12-week-old C57BL/6J mice. Murine pancreatic cancer cells (KPC) were orthotopically implanted into the pancreas of wild-type, IL-6-/-, and hepatocyte STAT3-/- male and female mice. Mice were subject to fasting, 50% food restriction, ad libitum feeding or ketogenic diet interventions. We measured longitudinal body composition by EchoMRI, body mass and food intake. At the endpoint, we measured tissue mass, tissue gene expression by quantitative real-time polymerase chain reaction, whole-body calorimetry, circulating hormone levels, faecal protein and lipid content, hepatic lipid content and ketogenic response to medium-chain fatty acid bolus. We assessed muscle atrophy in vivo and C2C12 myotube atrophy in vitro. RESULTS: Pre-cachectic PDAC mice did not preserve gastrocnemius muscle mass during 3-day food restriction (-13.1 ± 7.7% relative to food-restricted sham, P = 0.0117) and displayed impaired fatty acid oxidation during fasting, resulting in a hypoketotic state (ketogenic response to octanoate bolus, -83.0 ± 17.3%, P = 0.0328; Hmgcs2 expression, -28.3 ± 7.6%, P = 0.0004). PDAC human patients display impaired fasting ketones (-46.9 ± 7.1%, P < 0.0001) and elevated circulating interleukin-6 (IL-6) (12.4 ± 16.5-fold increase, P = 0.0001). IL-6-/- PDAC mice had improved muscle mass (+35.0 ± 3.9%, P = 0.0031) and ketogenic response (+129.4 ± 44.4%, P = 0.0033) relative to wild-type PDAC mice. Hepatocyte-specific signal transducer and activator of transcription 3 (STAT3) deletion prevented muscle loss (+9.3 ± 4.0%, P = 0.009) and improved fasting ketone levels (+52.0 ± 43.3%, P = 0.018) in PDAC mice. Without affecting tumour growth, a carbohydrate-free diet improved tibialis anterior myofibre diameter (+16.5 ± 3.5%, P = 0.0089), circulating ketone bodies (+333.0 ± 117.6%, P < 0.0001) and Hmgcs2 expression (+106.5 ± 36.1%, P < 0.0001) in PDAC mice. Ketone supplementation protected muscle against PDAC-induced atrophy in vitro (+111.0 ± 17.6%, P < 0.0001 myofibre diameter). CONCLUSIONS: In early PDAC cachexia, muscle vulnerability to wasting is dependent on inflammation-driven metabolic reprogramming in the liver. PDAC suppresses lipid ß-oxidation and impairs ketogenesis in the liver, which is reversed in genetically modified mouse models deficient in IL-6/STAT3 signalling or through ketogenic diet supplementation. This work establishes a direct link between skeletal muscle homeostasis and hepatic metabolism. Dietary and anti-inflammatory interventions that restore ketogenesis may be a viable preventative approach for pre-cachectic patients with pancreatic cancer.

2.
BMC Cancer ; 23(1): 1087, 2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37946117

ABSTRACT

BACKGROUND: Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating and dose-limiting side effect of systemic cancer therapy. In many cancer survivors, CIPN persists after treatment ends and is associated with functional impairments, abnormal gait patterns, falls, and diminished quality of life. However, little is known regarding which patients are most likely to develop CIPN symptoms that impair mobility and increase fall risk, when this risk develops, or the optimal timing of early intervention efforts to mitigate the impact of CIPN on functioning and fall risk. This study will address these knowledge gaps by (1) characterizing trajectories of symptoms, functioning, and falls before, during, and after treatment in adults prescribed neurotoxic chemotherapy for cancer; and (2) determining the simplest set of predictors for identifying individuals at risk for CIPN-related functional decline and falls. METHODS: We will enroll 200 participants into a prospective, observational study before initiating chemotherapy and up to 1 year after completing chemotherapy. Eligible participants are aged 40-85 years, diagnosed with stage I-III cancer, and scheduled to receive neurotoxic chemotherapy. We perform objective assessments of vibratory and touch sensation (biothesiometry, tuning fork, monofilament tests), standing and dynamic balance (quiet stance, Timed-Up-and-Go tests), and upper and lower extremity strength (handgrip dynamometry, 5-time repeated chair stand test) in the clinic at baseline, every 4-6 weeks during chemotherapy, and quarterly for 1 year post-chemotherapy. Participants wear devices that passively and continuously measure daily gait quality and physical activity for 1 week after each objective assessment and self-report symptoms (CIPN, insomnia, fatigue, dizziness, pain, cognition, anxiety, and depressive symptoms) and falls via weekly electronic surveys. We will use structural equation modeling, including growth mixture modeling, to examine patterns in trajectories of changes in symptoms, functioning, and falls associated with neurotoxic chemotherapy and then search for distinct risk profiles for CIPN. DISCUSSION: Identifying simple, early predictors of functional decline and fall risk in adults with cancer receiving neurotoxic chemotherapy will help identify individuals who would benefit from early and targeted interventions to prevent CIPN-related falls and disability. TRIAL REGISTRATION: This study was retrospectively registered with ClinicalTrials.gov (NCT05790538) on 3/30/2023.


Subject(s)
Antineoplastic Agents , Neoplasms , Neurotoxicity Syndromes , Peripheral Nervous System Diseases , Adult , Humans , Antineoplastic Agents/adverse effects , Hand Strength , Neoplasms/complications , Observational Studies as Topic , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/drug therapy , Prospective Studies , Quality of Life , Middle Aged , Aged , Aged, 80 and over
3.
Am J Surg ; 224(2): 742-746, 2022 08.
Article in English | MEDLINE | ID: mdl-35396132

ABSTRACT

BACKGROUND: Sarcopenia is associated with complications and inferior oncologic outcomes in solid tumors. Axial computed tomography (CT) scans can be used to evaluate sarcopenia, however manual quantification is laborious. We sought to validate an automated method of quantifying muscle cross-sectional area (CSA) in patients with pancreatic adenocarcinoma (PDAC). METHODS: Mid-L3 CT images from patients with PDAC were analyzed: CSAs of skeletal muscle (SM) were measured using manual segmentation and the software AutoMATiCA, and then compared with linear regression. RESULTS: Five-hundred-twenty-five unique scans were analyzed. There was robust correlation between manual and automated segmentation for L3 CSA (R2 0.94, P < 0.001). Bland-Altman analysis demonstrated a consistent overestimation of muscle CSA by AutoMATiCA with a mean difference of 5.7%. A correction factor of 1.06 was validated using a unique test dataset of 36 patients with non-PDAC peripancreatic malignancies. CONCLUSIONS: Automated muscle CSA measurement with AutoMATiCA is highly efficient and yields results highly correlated with manual measurement. These findings support the potential use of high-throughput sarcopenia analysis with abdominal CT scans for both clinical and research purposes.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Sarcopenia , Adenocarcinoma/complications , Adenocarcinoma/diagnostic imaging , Body Composition , Humans , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/diagnostic imaging , Sarcopenia/complications , Sarcopenia/diagnostic imaging , Tomography, X-Ray Computed/methods , Pancreatic Neoplasms
4.
Mol Metab ; 58: 101441, 2022 04.
Article in English | MEDLINE | ID: mdl-35031523

ABSTRACT

OBJECTIVE: Cancer cachexia is a devastating chronic condition characterized by involuntary weight loss, muscle wasting, abnormal fat metabolism, anorexia, and fatigue. However, the molecular mechanisms underlying this syndrome remain poorly understood. In particular, the hypothalamus may play a central role in cachexia, given that it has direct access to peripheral signals because of its anatomical location and attenuated blood-brain barrier. Furthermore, this region has a critical role in regulating appetite and metabolism. METHODS: To provide a detailed analysis of the hypothalamic response to cachexia, we performed single-cell RNA-seq combined with RNA-seq of the medial basal hypothalamus (MBH) in a mouse model for pancreatic cancer. RESULTS: We found many cell type-specific changes, such as inflamed endothelial cells, stressed oligodendrocyes and both inflammatory and moderating microglia. Lcn2, a newly discovered hunger suppressing hormone, was the highest induced gene. Interestingly, cerebral treatment with LCN2 not only induced many of the observed molecular changes in cachexia but also affected gene expression in food-intake decreasing POMC neurons. In addition, we found that many of the cachexia-induced molecular changes found in the hypothalamus mimic those at the primary tumor site. CONCLUSION: Our data reveal that multiple cell types in the MBH are affected by tumor-derived factors or host factors that are induced by tumor growth, leading to a marked change in the microenvironment of neurons critical for behavioral, metabolic, and neuroendocrine outputs dysregulated during cachexia. The mechanistic insights provided in this study explain many of the clinical features of cachexia and will be useful for future therapeutic development.


Subject(s)
Cachexia , Pancreatic Neoplasms , Animals , Cachexia/metabolism , Endothelial Cells/metabolism , Gene Regulatory Networks , Hypothalamus/metabolism , Mice , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Sequence Analysis, RNA , Tumor Microenvironment , Pancreatic Neoplasms
5.
Nat Commun ; 12(1): 2057, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33824339

ABSTRACT

Lipocalin 2 (LCN2) was recently identified as an endogenous ligand of the type 4 melanocortin receptor (MC4R), a critical regulator of appetite. However, it remains unknown if this molecule influences appetite during cancer cachexia, a devastating clinical entity characterized by decreased nutrition and progressive wasting. We demonstrate that LCN2 is robustly upregulated in murine models of pancreatic cancer, its expression is associated with reduced food consumption, and Lcn2 deletion is protective from cachexia-anorexia. Consistent with LCN2's proposed MC4R-dependent role in cancer-induced anorexia, pharmacologic MC4R antagonism mitigates cachexia-anorexia, while restoration of Lcn2 expression in the bone marrow is sufficient in restoring the anorexia feature of cachexia. Finally, we observe that LCN2 levels correlate with fat and lean mass wasting and is associated with increased mortality in patients with pancreatic cancer. Taken together, these findings implicate LCN2 as a pathologic mediator of appetite suppression during pancreatic cancer cachexia.


Subject(s)
Appetite , Cachexia/complications , Lipocalin-2/metabolism , Pancreatic Neoplasms/complications , Adult , Aged , Aged, 80 and over , Animals , Anorexia/blood , Anorexia/complications , Blood-Brain Barrier/pathology , Bone Marrow/pathology , Cachexia/blood , Cell Line, Tumor , Disease Models, Animal , Feeding Behavior , Female , Gene Deletion , Humans , Lipocalin-2/blood , Male , Mice, Knockout , Middle Aged , Models, Biological , Muscles/pathology , Neutrophils/pathology , Organ Size , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/metabolism , Up-Regulation
6.
JAMA Otolaryngol Head Neck Surg ; 146(8): 714-722, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32525518

ABSTRACT

Importance: The negative association of low lean muscle mass (sarcopenia) with survival outcomes in head and neck cancers, including oropharyngeal carcinoma, is established. However, it is not known whether the choice of primary treatment modality (surgery or radiotherapy) is associated with oncologic outcomes of patients with sarcopenia and oropharyngeal squamous cell carcinoma (OPSCC). Objective: To examine whether primary surgical resection or definitive radiotherapy is associated with improved survival for patients with sarcopenia and localized OPSCC. Design, Setting, and Participants: A cohort study was conducted of patients with clinically staged T1 to T2, N0 to N2 OPSCC with cross-sectional abdominal imaging within 60 days prior to treatment and treated between January 1, 2005, and December 31, 2017. Skeletal muscle mass was measured at the third lumbar vertebra using previously defined techniques and sarcopenia was defined as less than 52.4 cm2/m2 of muscle for men and less than 38.5 cm2/m2 for women. In addition, associated patient demographic characteristics, cancer data, treatment information, and survival outcomes were assessed. Statistical analysis was performed from December 3, 2018, to August 28, 2019. Main Outcomes and Measures: Primary outcomes were overall survival and disease-specific survival. Results: Among the 245 patients who met study inclusion criteria, 209 were men (85.3%) and the mean (SD) age was 62.3 (7.8) years. Sarcopenia was detected in 135 patients (55.1%), while normal skeletal muscle mass was detected in 110 patients (44.9%). For the 110 patients without sarcopenia, primary treatment modality was not associated with improved survival. For patients with sarcopenia at diagnosis, primary surgical resection was associated with improved overall survival (hazard ratio [HR], 0.37; 95% CI, 0.17-0.82) and disease-specific survival (HR, 0.22; 95% CI, 0.07-0.68). This association persisted after propensity score matching, as up-front surgery was associated with improved overall survival (HR, 0.33; 95% CI, 0.12-0.91) and disease-specific survival (HR, 0.17; 95% CI, 0.04-0.75) survival. Conclusions and Relevance: This study suggests that sarcopenia has a negative association with survival for patients with OPSCC. Primary surgery and radiotherapy confer similar survival associations for patients with normal skeletal muscle mass and localized OPSCC. However, up-front surgical resection may be associated with improved survival outcomes for patients with sarcopenia.


Subject(s)
Head and Neck Neoplasms/therapy , Propensity Score , Robotic Surgical Procedures/methods , Sarcopenia/etiology , Squamous Cell Carcinoma of Head and Neck/therapy , Aged , Cross-Sectional Studies , Female , Follow-Up Studies , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/diagnosis , Humans , Incidence , Male , Middle Aged , Neoplasm Staging , Oregon/epidemiology , Retrospective Studies , Sarcopenia/diagnosis , Sarcopenia/epidemiology , Squamous Cell Carcinoma of Head and Neck/complications , Squamous Cell Carcinoma of Head and Neck/diagnosis , Survival Rate/trends
7.
JAMA Otolaryngol Head Neck Surg ; 145(7): 647-654, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31169874

ABSTRACT

Importance: Sarcopenia, or the loss of muscle mass, is associated with poor treatment outcomes in a variety of surgical fields. However, the association between sarcopenia and long-term survival in a broad cohort of patients with head and neck cancer (HNC) is unknown. Objective: To determine whether sarcopenia is associated with long-term survival in patients undergoing major head and neck surgery for HNC. Design, Setting, and Participants: A retrospective medical records review was conducted at a tertiary care academic hospital. Two hundred sixty patients undergoing major head and neck ablative procedures with cross-sectional abdominal imaging performed within 45 days prior to surgery were included in the analysis. The study was conducted from January 1, 2005, to December 31, 2016. Data analysis was performed from June 1, 2018, to February 28, 2019. Interventions: Measurement of cross-sectional muscle area at the L3 vertebra level. Main Outcomes and Measures: Two- and 5-year overall survival were the primary outcomes. Results: Of the 260 patients included in the study, 193 were men (74.2%); mean (SD) age was 61.1 (11) years. Sarcopenia was present in 144 patients (55.4%). Two-year overall survival was 71.9% of the patients (n = 82) in the sarcopenia group compared with 88.5% of the patients (n = 85) in the nonsarcopenia group (odds ratio [OR], 0.33; 95% CI, 0.16-0.70). At 5 years, overall survival was 36.5% in patients (n = 23) with sarcopenia and 60.5% in patients (n = 26) without sarcopenia (OR, 0.38; 95% CI, 0.17-0.84). On multivariate analysis, sarcopenia was a significant negative predictor of both 2-year (OR, 0.33; 95% CI, 0.14-0.77) and 5-year (OR, 0.38; 95% CI, 0.17-0.84) overall survival. Conclusions and Relevance: Sarcopenia appears to be a significant negative predictor of long-term overall survival in patients with HNC undergoing major head and neck surgery. Sarcopenia may be accurately assessed on cross-sectional imaging and may be useful clinically as a prognostic variable and as an area for intervention to improve treatment outcomes.


Subject(s)
Head and Neck Neoplasms/mortality , Sarcopenia/mortality , Body Composition/physiology , Body Mass Index , Disease-Free Survival , Female , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/surgery , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Oregon/epidemiology , Retrospective Studies , Risk Factors , Sarcopenia/complications
8.
Theranostics ; 8(19): 5276-5288, 2018.
Article in English | MEDLINE | ID: mdl-30555546

ABSTRACT

Muscle atrophy occurs during chronic diseases, resulting in diminished quality of life and compromised treatment outcomes. There is a high demand for therapeutics that increase muscle mass while abrogating the need for special dietary and exercise requirements. Therefore, we developed an efficient nanomedicine approach capable of increasing muscle mass. Methods: The therapy is based on nanoparticle-mediated delivery of follistatin messenger RNA (mRNA) to the liver after subcutaneous administration. The delivered mRNA directs hepatic cellular machinery to produce follistatin, a glycoprotein that increases lean mass through inhibition of negative regulators of muscle mass (myostatin and activin A). These factors are elevated in numerous disease states, thereby providing a target for therapeutic intervention. Results: Animal studies validated that mRNA-loaded nanoparticles enter systemic circulation following subcutaneous injection, accumulate and internalize in the liver, where the mRNA is translated into follistatin. Follistatin serum levels were elevated for 72 h post injection and efficiently reduced activin A and myostatin serum concentrations. After eight weeks of repeated injections, the lean mass of mice in the treatment group was ~10% higher when compared to that of the controls. Conclusion: Based on the obtained results demonstrating an increased muscle mass as well as restricted fat accumulation, this nanoplatform might be a milestone in the development of mRNA technologies and the treatment of muscle wasting disorders.


Subject(s)
Drug Carriers/administration & dosage , Follistatin/genetics , Liver/metabolism , Muscle Development/drug effects , Nanoparticles/administration & dosage , RNA, Messenger/administration & dosage , Animals , Injections, Subcutaneous , Mice , Treatment Outcome
9.
Mol Cell ; 72(6): 985-998.e7, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30415949

ABSTRACT

Current models of SIRT1 enzymatic regulation primarily consider the effects of fluctuating levels of its co-substrate NAD+, which binds to the stably folded catalytic domain. By contrast, the roles of the sizeable disordered N- and C-terminal regions of SIRT1 are largely unexplored. Here we identify an insulin-responsive sensor in the SIRT1 N-terminal region (NTR), comprising an acidic cluster (AC) and a 3-helix bundle (3HB), controlling deacetylase activity. The allosteric assistor DBC1 removes a distal N-terminal shield from the 3-helix bundle, permitting PACS-2 to engage the acidic cluster and the transiently exposed helix 3 of the 3-helix bundle, disrupting its structure and inhibiting catalysis. The SIRT1 activator (STAC) SRT1720 binds and stabilizes the 3-helix bundle, protecting SIRT1 from inhibition by PACS-2. Identification of the SIRT1 insulin-responsive sensor and its engagement by the DBC1 and PACS-2 regulatory hub provides important insight into the roles of disordered regions in enzyme regulation and the mode by which STACs promote metabolic fitness.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hepatocytes/enzymology , Insulin/metabolism , Sirtuin 1/metabolism , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Allosteric Regulation , Animals , Binding Sites , Diet, High-Fat , Disease Models, Animal , Gene Expression Regulation , HCT116 Cells , Hepatocytes/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/genetics , Obesity/prevention & control , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Sirtuin 1/genetics , Vesicular Transport Proteins/deficiency , Vesicular Transport Proteins/genetics
10.
J Neuroinflammation ; 14(1): 219, 2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29121947

ABSTRACT

BACKGROUND: The physiological and behavioral symptoms of sickness, including fever, anorexia, behavioral depression, and weight loss can be both beneficial and detrimental. These sickness responses are triggered by pro-inflammatory cytokines acting on cells within the brain. Previous research demonstrates that the febrile response to peripheral insults depends upon prostaglandin production by vascular endothelial cells, but the mechanisms and specific cell type(s) responsible for other sickness responses remain unknown. The purpose of the present study was to identify which cells within the brain are required for sickness responses triggered by central nervous system inflammation. METHODS: Intracerebroventricular (ICV) administration of 10 ng of the potent pro-inflammatory cytokine interleukin-1ß (IL-1ß) was used as an experimental model of central nervous system cytokine production. We examined which cells respond to IL-1ß in vivo via fluorescent immunohistochemistry. Using multiple transgenic mouse lines expressing Cre recombinase under the control of cell-specific promoters, we eliminated IL-1ß signaling from different populations of cells. Food consumption, body weight, movement, and temperature were recorded in adult male mice and analyzed by two-factor ANOVA to determine where IL-1ß signaling is essential for sickness responses. RESULTS: Endothelial cells, microglia, ependymal cells, and astrocytes exhibit nuclear translocation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) in response to IL-1ß. Interfering with IL-1ß signaling in microglia, endothelial cells within the parenchyma of the brain, or both did not affect sickness responses. Only mice that lacked IL-1ß signaling in all endothelium including fenestrated capillaries lacked sickness responses. CONCLUSIONS: These experiments show that IL-1ß-induced sickness responses depend on intact IL-1ß signaling in blood vessels and suggest that fenestrated capillaries act as a critical signaling relay between the immune and nervous systems. TRIAL REGISTRATION: Not applicable.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/metabolism , Illness Behavior/drug effects , Inflammation/pathology , Interleukin-1beta/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Capillaries/drug effects , Capillaries/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Illness Behavior/physiology , Inflammation/metabolism , Interleukin-1beta/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/drug effects , Signal Transduction/physiology
11.
J Neuroinflammation ; 14(1): 133, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28668091

ABSTRACT

BACKGROUND: During acute infections and chronic illnesses, the pro-inflammatory cytokine interleukin-1ß (IL-1ß) acts within the brain to elicit metabolic derangements and sickness behaviors. It is unknown which cells in the brain are the proximal targets for IL-1ß with respect to the generation of these illness responses. We performed a series of in vitro experiments to (1) investigate which brain cell populations exhibit inflammatory responses to IL-1ß and (2) examine the interactions between different IL-1ß-responsive cell types in various co-culture combinations. METHODS: We treated primary cultures of murine brain microvessel endothelial cells (BMEC), astrocytes, and microglia with PBS or IL-1ß, and then performed qPCR to measure inflammatory gene expression or immunocytochemistry to evaluate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. To evaluate whether astrocytes and/or BMEC propagate inflammatory signals to microglia, we exposed microglia to astrocyte-conditioned media and co-cultured endothelial cells and glia in transwells. Treatment groups were compared by Student's t tests or by ANOVA followed by Bonferroni-corrected t tests. RESULTS: IL-1ß increased inflammatory gene expression and NF-κB activation in primary murine-mixed glia, enriched astrocyte, and BMEC cultures. Although IL-1ß elicited minimal changes in inflammatory gene expression and did not induce the nuclear translocation of NF-κB in isolated microglia, these cells were more robustly activated by IL-1ß when co-cultured with astrocytes and/or BMEC. We observed a polarized endothelial response to IL-1ß, because the application of IL-1ß to the abluminal endothelial surface produced a more complex microglial inflammatory response than that which occurred following luminal IL-1ß exposure. CONCLUSIONS: Inflammatory signals are detected, amplified, and propagated through the CNS via a sequential and reverberating signaling cascade involving communication between brain endothelial cells and glia. We propose that the brain's innate immune response differs depending upon which side of the blood-brain barrier the inflammatory stimulus arises, thus allowing the brain to respond differently to central vs. peripheral inflammatory insults.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Interleukin-1beta/pharmacology , Neuroglia/metabolism , Signal Transduction/physiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/blood supply , Brain/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Microvessels/metabolism , Neuroglia/drug effects , Signal Transduction/drug effects
12.
J Cachexia Sarcopenia Muscle ; 8(5): 824-838, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28730707

ABSTRACT

BACKGROUND: Cachexia is a complex metabolic and behavioural syndrome lacking effective therapies. Pancreatic ductal adenocarcinoma (PDAC) is one of the most important conditions associated with cachexia, with >80% of PDAC patients suffering from the condition. To establish the cardinal features of a murine model of PDAC-associated cachexia, we characterized the effects of implanting a pancreatic tumour cell line from a syngeneic C57BL/6 KRASG12D P53R172H Pdx-Cre+/+ (KPC) mouse. METHODS: Male and female C57BL/6 mice were inoculated subcutaneously, intraperitoneally, or orthotopically with KPC tumour cells. We performed rigorous phenotypic, metabolic, and behavioural analysis of animals over the course of tumour development. RESULTS: All routes of administration produced rapidly growing tumours histologically consistent with moderate to poorly differentiated PDAC. The phenotype of this model was dependent on route of administration, with orthotopic and intraperitoneal implantation inducing more severe cachexia than subcutaneous implantation. KPC tumour growth decreased food intake, decreased adiposity and lean body mass, and decreased locomotor activity. Muscle catabolism was observed in both skeletal and cardiac muscles, but the dominant catabolic pathway differed between these tissues. The wasting syndrome in this model was accompanied by hypothalamic inflammation, progressively decreasing brown and white adipose tissue uncoupling protein 1 (Ucp1) expression, and increased peripheral inflammation. Haematological and endocrine abnormalities included neutrophil-dominant leukocytosis and anaemia, and decreased serum testosterone. CONCLUSIONS: Syngeneic KPC allografts are a robust model for studying cachexia, which recapitulate key features of the PDAC disease process and induce a wide array of cachexia manifestations. This model is therefore ideally suited for future studies exploring the physiological systems involved in cachexia and for preclinical studies of novel therapies.


Subject(s)
Cachexia/etiology , Cachexia/pathology , Pancreatic Neoplasms/complications , Allografts , Anemia/etiology , Anemia/metabolism , Anemia/pathology , Animals , Biopsy , Body Composition , Cachexia/diagnostic imaging , Disease Models, Animal , Energy Metabolism , Female , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Leukocytosis , Locomotion , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Muscles/metabolism , Muscles/pathology , Neutrophil Infiltration , Testosterone/metabolism
13.
PLoS One ; 11(12): e0168731, 2016.
Article in English | MEDLINE | ID: mdl-27997622

ABSTRACT

BACKGROUND: Steroid-induced sleep disturbance is a common and highly distressing morbidity for children receiving steroid chemotherapy for the treatment of pediatric acute lymphoblastic leukemia (ALL). Sleep disturbance can negatively impact overall quality of life, neurodevelopment, memory consolidation, and wound healing. Hypothalamic orexin neurons are influential wake-promoting neurons, and disturbances in orexin signaling leads to abnormal sleep behavior. A new class of drug, the orexin receptor antagonists, could be an intriguing option for sleep disorders caused by increased orexinergic output. Our aim was to examine the impact of ALL treatment doses of corticosteroids on the orexin system in rodents and in children undergoing treatment for childhood ALL. METHODS: We administered repeated injections of dexamethasone to rodents and measured responsive orexin neural activity compared to controls. In children with newly diagnosed standard risk B-cell ALL receiving dexamethasone therapy per Children's Oncology Group (COG) induction therapy from 2014-2016, we collected pre- and during-steroids matched CSF samples and measured the impact of steroids on CSF orexin concentration. RESULTS: In both rodents, all markers orexin signaling, including orexin neural output and orexin receptor expression, were preserved in the setting of dexamethasone. Additionally, we did not detect a difference in pre- and during-dexamethasone CSF orexin concentrations in children receiving dexamethasone. CONCLUSIONS: Our results demonstrate that rodent and human orexin physiology is largely preserved in the setting of high dose dexamethasone. The data obtained in our experimental model fail to demonstrate a causative role for disruption of the orexin pathway in steroid-induced sleep disturbance.


Subject(s)
Dexamethasone , Hypothalamus , Neurons/metabolism , Orexins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Signal Transduction/drug effects , Adolescent , Animals , Child , Child, Preschool , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Female , Humans , Hypothalamus/metabolism , Hypothalamus/physiopathology , Male , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Rats , Rats, Sprague-Dawley , Sleep Wake Disorders/chemically induced , Sleep Wake Disorders/metabolism , Sleep Wake Disorders/physiopathology
14.
Mol Metab ; 4(1): 25-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25685687

ABSTRACT

OBJECTIVE: Recent evidence indicates that the adult hematopoietic system is susceptible to diet-induced lineage skewing. It is not known whether the developing hematopoietic system is subject to metabolic programming via in utero high-fat diet (HFD) exposure, an established mechanism of adult disease in several organ systems. We previously reported substantial losses in offspring liver size with prenatal HFD. As the liver is the main hematopoietic organ in the fetus, we asked whether the developmental expansion of the hematopoietic stem and progenitor cell (HSPC) pool is compromised by prenatal HFD and/or maternal obesity. METHODS: We used quantitative assays, progenitor colony formation, flow cytometry, transplantation, and gene expression assays with a series of dietary manipulations to test the effects of gestational high-fat diet and maternal obesity on the day 14.5 fetal liver hematopoietic system. RESULTS: Maternal obesity, particularly when paired with gestational HFD, restricts physiological expansion of fetal HSPCs while promoting the opposing cell fate of differentiation. Importantly, these effects are only partially ameliorated by gestational dietary adjustments for obese dams. Competitive transplantation reveals compromised repopulation and myeloid-biased differentiation of HFD-programmed HSPCs to be a niche-dependent defect, apparent in HFD-conditioned male recipients. Fetal HSPC deficiencies coincide with perturbations in genes regulating metabolism, immune and inflammatory processes, and stress response, along with downregulation of genes critical for hematopoietic stem cell self-renewal and activation of pathways regulating cell migration. CONCLUSIONS: Our data reveal a previously unrecognized susceptibility to nutritional and metabolic developmental programming in the fetal HSPC compartment, which is a partially reversible and microenvironment-dependent defect perturbing stem and progenitor cell expansion and hematopoietic lineage commitment.

15.
PLoS One ; 9(5): e97529, 2014.
Article in English | MEDLINE | ID: mdl-24824753

ABSTRACT

Inhibition of soluble epoxide hydrolase (sEH) is a potential target of therapy for ischemic injury. sEH metabolizes neuroprotective epoxyeicosatrienoic acids (EETs). We recently demonstrated that sEH inhibition reduces infarct size after middle cerebral artery occlusion (MCAO) in type 1 diabetic mice. We hypothesized that inhibition of sEH would protect against ischemic injury in type 2 diabetic mice. Type 2 diabetes was produced by combined high-fat diet, nicotinamide and streptozotocin in male mice. Diabetic and control mice were treated with vehicle or the sEH inhibitor t-AUCB then subjected to 60-min MCAO. Compared to chow-fed mice, high fat diet-fed mice exhibited an upregulation of sEH mRNA and protein in brain, but no differences in brain EETs levels were observed between groups. Type 2 diabetic mice had increased blood glucose levels at baseline and throughout ischemia, decreased laser-Doppler perfusion of the MCA territory after reperfusion, and sustained larger cortical infarcts compared to control mice. t-AUCB decreased fasting glucose levels at baseline and throughout ischemia, improved cortical perfusion after MCAO and significantly reduced infarct size in diabetic mice. We conclude that sEH inhibition, as a preventative treatment, improves glycemic status, post-ischemic reperfusion in the ischemic territory, and stroke outcome in type 2 diabetic mice.


Subject(s)
Benzoates/pharmacology , Diabetes Mellitus, Experimental/complications , Epoxide Hydrolases/antagonists & inhibitors , Stroke/etiology , Stroke/prevention & control , Urea/analogs & derivatives , Analysis of Variance , Animals , Blood Glucose/metabolism , Blotting, Western , Brain/metabolism , Diet, High-Fat , Epoxide Hydrolases/metabolism , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Niacinamide , Real-Time Polymerase Chain Reaction , Urea/pharmacology
16.
J Cereb Blood Flow Metab ; 33(10): 1650-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23899929

ABSTRACT

Hyperglycemia worsens stroke, yet rigorous glycemic control does not improve neurologic outcome. An alternative is to target downstream molecular mediator(s) triggered by hyperglycemia but independent of prevailing glycemia. Soluble epoxide hydrolase (sEH) is a potential mediator of injury via its metabolism of neuroprotective epoxyeicosatrienoic acids (EETs). We tested whether hyperglycemia exacerbates cerebral injury by upregulating sEH and decreasing brain EET levels. Type 1 diabetes mellitus was modeled by streptozotocin (STZ; 50 mg/kg per day intraperitoneally, 5 days) in male mice. At 4 weeks, STZ-treated and control mice underwent 45-minute middle cerebral artery occlusion (MCAO) with or without sEH blockade by trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB; 1 mg/kg intraperitoneally daily for 6 days before MCAO). The STZ-treated mice had increased sEH mRNA expression in cerebral vessels and decreased EET concentrations in brain. There was no difference in cortical perfusion between groups. The STZ-treated mice sustained larger brain infarct than controls. Pretreatment with t-AUCB eliminated the difference in infarct size and EETs concentration between STZ-treated mice and controls, without altering glycemia. We conclude that type 1 diabetes mellitus upregulates sEH mRNA and decreases concentrations of neuroprotective EETs within the brain, leading to worse stroke outcome. The data indicate that sEH antagonism may be beneficial in the setting of hyperglycemic stroke.


Subject(s)
8,11,14-Eicosatrienoic Acid/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/enzymology , Epoxide Hydrolases/biosynthesis , Stroke/enzymology , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , Animals , Benzoates/pharmacology , Blood Glucose/analysis , Cerebral Angiography , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Epoxide Hydrolases/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Optical Imaging , Real-Time Polymerase Chain Reaction , Streptozocin/pharmacology , Stroke/etiology , Stroke/prevention & control , Up-Regulation , Urea/analogs & derivatives , Urea/pharmacology
17.
FASEB J ; 27(9): 3572-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23733748

ABSTRACT

Cachexia is a wasting condition defined by skeletal muscle atrophy in the setting of systemic inflammation. To explore the site at which inflammatory mediators act to produce atrophy in vivo, we utilized mice with a conditional deletion of the inflammatory adaptor protein myeloid differentiation factor 88 (MyD88). Although whole-body MyD88-knockout (wbMyD88KO) mice resist skeletal muscle atrophy in response to LPS, muscle-specific deletion of MyD88 is not protective. Furthermore, selective reexpression of MyD88 in the muscle of wbMyD88KO mice via electroporation fails to restore atrophy gene induction by LPS. To evaluate the role of glucocorticoids as the inflammation-induced mediator of atrophy in vivo, we generated mice with targeted deletion of the glucocorticoid receptor in muscle (mGRKO mice). Muscle-specific deletion of the glucocorticoid receptor affords a 71% protection against LPS-induced atrophy compared to control animals. Furthermore, mGRKO mice exhibit 77% less skeletal muscle atrophy than control animals in response to tumor growth. These data demonstrate that glucocorticoids are a major determinant of inflammation-induced atrophy in vivo and play a critical role in the pathogenesis of endotoxemic and cancer cachexia.


Subject(s)
Cachexia/etiology , Cachexia/metabolism , Carcinoma, Lewis Lung/physiopathology , Glucocorticoids/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Myeloid Differentiation Factor 88/metabolism , Animals , Blotting, Western , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Immunohistochemistry , In Situ Hybridization , Inflammation Mediators/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/genetics , Myeloid Differentiation Factor 88/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Am J Physiol Endocrinol Metab ; 303(12): E1446-58, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23047987

ABSTRACT

Animals exhibit a rapid and sustained anorexia when fed a diet that is deficient in a single indispensable amino acid (IAA). The chemosensor for IAA deficiency resides within the anterior piriform cortex (APC). Although the cellular and molecular mechanisms by which the APC detects IAA deficiency are well established, the efferent neural pathways that reduce feeding in response to an IAA-deficient diet remain to be fully characterized. In the present work, we investigated whether 1) central melanocortin signaling is involved in IAA deficiency-induced anorexia (IAADA) and 2) IAADA engages other key appetite-regulating neuronal populations in the hypothalamus. Rats and mice that consumed a valine-deficient diet (VDD) for 2-3 wk exhibited marked reductions in food intake, body weight, fat and lean body mass, body temperature, and white adipose tissue leptin gene expression, as well as a paradoxical increase in brown adipose tissue uncoupling protein-1 mRNA. Animals consuming the VDD had altered hypothalamic gene expression, typical of starvation. Pharmacological and genetic blockade of central melanocortin signaling failed to increase long-term food intake in this model. Chronic IAA deficiency was associated with a marked upregulation of corticotropin-releasing hormone expression in the lateral hypothalamus, particularly in the parasubthalamic nucleus, an area heavily innervated by efferent projections from the APC. Our observations indicate that the hypothalamic melanocortin system plays a minor role in acute, but not chronic, IAADA and suggest that the restraint on feeding is analogous to that observed after chronic dehydration.


Subject(s)
Anorexia/etiology , Anorexia/metabolism , Hypothalamus/metabolism , Neural Pathways/metabolism , Neurons/metabolism , Signal Transduction , Valine/deficiency , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Anorexia/pathology , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Female , Gene Expression Regulation , Hypothalamus/pathology , Ion Channels/genetics , Ion Channels/metabolism , Leptin/genetics , Leptin/metabolism , Male , Melanocortins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neural Pathways/pathology , Neurons/pathology , Organ Specificity , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Uncoupling Protein 1 , Valine/metabolism
19.
Am J Physiol Endocrinol Metab ; 301(6): E1243-53, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21900122

ABSTRACT

Maternal overnutrition prior to and during gestation causes pronounced metabolic dysfunction in the adult offspring. However, less is known about metabolic adaptations in the offspring that occur independently of postnatal growth and nutrition. Therefore, we evaluated the impact of excess maternal dietary lipid intake on the in utero programming of body composition, hepatic function, and hypothalamic development in newborn (P0) offspring. Female mice were fed a low-fat (LF) or high-fat (HF) diet and were mated after 4, 12, and 23 wk. A subset of the obese HF dams was switched to the LF diet during the second (DR2) or third (DR3) pregnancies. The HF offspring accrued more fat mass than the LF pups, regardless of duration of maternal HF diet consumption or prepregnancy maternal adiposity. Increased neonatal adiposity was not observed in the DR3 pups. Liver weights were reduced in the HF offspring but not in the DR2 or DR3 pups. Offspring hepatic triglyceride content was reduced in the HF pups, but hepatic inflammation and expression of lipid metabolism genes were largely unaffected by maternal diet. Maternal diet did not alter the hypothalamic expression of orexigenic and anorexigenic neuropeptides in the offspring. Thus, the intrauterine programming of increased neonatal adiposity and reduced liver size by maternal overnutrition is evident in mice at birth and occurs prior to the development of maternal obesity. These observations demonstrate that dietary intervention during pregnancy minimizes the deleterious effects of maternal obesity on offspring body composition, potentially reducing the offsprings' risk of developing obesity and related diseases later in life.


Subject(s)
Body Composition/drug effects , Dietary Fats/pharmacology , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects/metabolism , Algorithms , Animals , Animals, Newborn , Diet, High-Fat , Eating/drug effects , Eating/physiology , Female , Male , Maternal Nutritional Physiological Phenomena/drug effects , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/physiopathology , Up-Regulation
20.
Curr Opin Support Palliat Care ; 4(4): 266-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20733497

ABSTRACT

PURPOSE OF REVIEW: Cachexia occurs in various inflammatory diseases and is characterized by weight loss and muscle wasting. Pro-inflammatory cytokines modulate the activity of neuropeptides and hormones that control energy homeostasis and/or illness behaviors. This review summarizes recent (published within the past 18 months) literature regarding neuropeptides and hormones that have been implicated in the pathophysiology of cachexia, and that are likely to have therapeutic potential for preventing or reversing cachexia in various disease states. RECENT FINDINGS: Hypothalamic pro-opiomelanocortin (POMC) and agouti-related protein (AgRP) neurons are downstream targets for pro-inflammatory cytokines. Genetic or pharmacological blockade of melanocortin receptor signaling preserves lean body mass and attenuates anorexia in experimental models of cachexia. Orally available melanocortin receptor antagonists have been developed and tested in cachectic animals with favorable results. Ghrelin and ghrelin mimetics increase appetite and preserve lean body mass in cachectic patients with diverse underlying diseases. Additional neuropeptide-expressing neurons in the hypothalamus (e.g., orexin neurons) might play a role in cachexia-associated lethargy. SUMMARY: Promising outcomes from recent preclinical studies and/or early clinical trials with melanocortin receptor antagonists and ghrelin mimetics raise hopes that safe and effective anti-cachexia drugs will soon become available for widespread clinical use.


Subject(s)
Cachexia/drug therapy , Cachexia/metabolism , Neuropeptides/metabolism , Neuropeptides/therapeutic use , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...