Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 637
Filtrer
1.
Int J Surg Oncol ; 2024: 5339292, 2024.
Article de Anglais | MEDLINE | ID: mdl-38966634

RÉSUMÉ

Objective: Determine the histopathologic features that correlate with head and neck cancer (HNC) cachexia. Methods: A single-institution, retrospective study was performed on adults with HPV-negative, mucosal squamous cell carcinoma of the aerodigestive tract undergoing resection and free flap reconstruction from 2014 to 2019. Patients with distant metastases were excluded. Demographics, comorbidities, preoperative nutrition, and surgical pathology reports were collected. Comparisons of histopathologic features and cachexia severity were made. Results: The study included 222 predominantly male (64.9%) patients aged 61.3 ± 11.8 years. Cachexia was identified in 57.2% patients, and 18.5% were severe (≥15% weight loss). No differences in demographics were identified between the groups. Compared to control, patients with severe cachexia had lower serum hemoglobin (p=0.048) and albumin (p < 0.001), larger tumor diameter (p < 0.001), greater depth of invasion (p < 0.001), and elevated proportions of pT4 disease (p < 0.001), pN2-N3 disease (p=0.001), lymphovascular invasion (p=0.009), and extranodal extension (p=0.014). Multivariate logistic regression identified tumor size (OR [95% CI] = 1.36 [1.08-1.73]), oral cavity tumor (OR [95% CI] = 0.30 [0.11-0.84]), and nodal burden (OR [95% CI] = 1.16 [0.98-1.38]) as significant histopathologic contributors of cancer cachexia. Conclusions: Larger, more invasive tumors with nodal metastases and aggressive histologic features are associated with greater cachexia severity in mucosal HNC.


Sujet(s)
Cachexie , Tumeurs de la tête et du cou , Humains , Cachexie/anatomopathologie , Cachexie/étiologie , Mâle , Adulte d'âge moyen , Femelle , Études rétrospectives , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/chirurgie , Tumeurs de la tête et du cou/complications , Sujet âgé , Carcinome épidermoïde de la tête et du cou/chirurgie , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/complications , Pronostic , Invasion tumorale , Lambeaux tissulaires libres
2.
Physiol Rep ; 12(13): e16103, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38946587

RÉSUMÉ

Cancer cachexia is a multifactorial syndrome associated with advanced cancer that contributes to mortality. Cachexia is characterized by loss of body weight and muscle atrophy. Increased skeletal muscle mitochondrial reactive oxygen species (ROS) is a contributing factor to loss of muscle mass in cachectic patients. Mice inoculated with Lewis lung carcinoma (LLC) cells lose weight, muscle mass, and have lower muscle sirtuin-1 (sirt1) expression. Nicotinic acid (NA) is a precursor to nicotinamide dinucleotide (NAD+) which is exhausted in cachectic muscle and is a direct activator of sirt1. Mice lost body and muscle weight and exhibited reduced skeletal muscle sirt1 expression after inoculation with LLC cells. C2C12 myotubes treated with LLC-conditioned media (LCM) had lower myotube diameter. We treated C2C12 myotubes with LCM for 24 h with or without NA for 24 h. C2C12 myotubes treated with NA maintained myotube diameter, sirt1 expression, and had lower mitochondrial superoxide. We then used a sirt1-specific small molecule activator SRT1720 to increase sirt1 activity. C2C12 myotubes treated with SRT1720 maintained myotube diameter, prevented loss of sirt1 expression, and attenuated mitochondrial superoxide production. Our data provides evidence that NA may be beneficial in combating cancer cachexia by maintaining sirt1 expression and decreasing mitochondrial superoxide production.


Sujet(s)
Cachexie , Fibres musculaires squelettiques , Stress oxydatif , Sirtuine-1 , Animaux , Cachexie/étiologie , Cachexie/métabolisme , Cachexie/anatomopathologie , Cachexie/prévention et contrôle , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/anatomopathologie , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Carcinome pulmonaire de Lewis/métabolisme , Carcinome pulmonaire de Lewis/anatomopathologie , Carcinome pulmonaire de Lewis/complications , Mâle , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Mitochondries du muscle/métabolisme , Mitochondries du muscle/effets des médicaments et des substances chimiques , Mitochondries du muscle/anatomopathologie , Lignée cellulaire , Acide nicotinique/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme
3.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38731953

RÉSUMÉ

Cardiac disorders in cancer patients pose significant challenges to disease prognosis. While it has been established that these disorders are linked to cancer cells, the precise underlying mechanisms remain elusive. In this study, we investigated the impact of cancerous ascites from the rat colonic carcinoma cell line RCN9 on H9c2 cardiomyoblast cells. We found that the ascites reduced mitochondrial volume, increased oxidative stress, and decreased membrane potential in the cardiomyoblast cells, leading to apoptosis and autophagy. Although the ascites fluid contained a substantial amount of high-mobility group box-1 (HMGB1), we observed that neutralizing HMGB1 with a specific antibody mitigated the damage inflicted on myocardial cells. Our mechanistic investigations revealed that HMGB1 activated both nuclear factor κB and phosphoinositide 3-kinases-AKT signals through HMGB1 receptors, namely the receptor for advanced glycation end products and toll-like receptor-4, thereby promoting apoptosis and autophagy. In contrast, treatment with berberine (BBR) induced the expression of miR-181c-5p and miR-340-5p while suppressing HMGB1 expression in RCN9 cells. Furthermore, BBR reduced HMGB1 receptor expression in cardiomyocytes, consequently mitigating HMGB1-induced damage. We validated the myocardial protective effects of BBR in a cachectic rat model. These findings underscore the strong association between HMGB1 and cancer cachexia, highlighting BBR as a promising therapeutic agent for myocardial protection through HMGB1 suppression and modulation of the signaling system.


Sujet(s)
Berbérine , Cachexie , Protéine HMGB1 , Animaux , Rats , Apoptose/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Berbérine/pharmacologie , Cachexie/métabolisme , Cachexie/traitement médicamenteux , Cachexie/étiologie , Cachexie/anatomopathologie , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Protéine HMGB1/effets des médicaments et des substances chimiques , Protéine HMGB1/métabolisme , microARN/génétique , microARN/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Tumeurs/métabolisme , Tumeurs/complications , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Rat Sprague-Dawley , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme
4.
Cells ; 13(9)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38727319

RÉSUMÉ

In our previous studies, we showed that the generation of ovarian tumors in NSG mice (immune-compromised) resulted in the induction of muscle and cardiac cachexia, and treatment with withaferin A (WFA; a steroidal lactone) attenuated both muscle and cardiac cachexia. However, our studies could not address if these restorations by WFA were mediated by its anti-tumorigenic properties that might, in turn, reduce the tumor burden or WFA's direct, inherent anti-cachectic properties. To address this important issue, in our present study, we used a cachectic model induced by the continuous infusion of Ang II by implanting osmotic pumps in immunocompetent C57BL/6 mice. The continuous infusion of Ang II resulted in the loss of the normal functions of the left ventricle (LV) (both systolic and diastolic), including a significant reduction in fractional shortening, an increase in heart weight and LV wall thickness, and the development of cardiac hypertrophy. The infusion of Ang II also resulted in the development of cardiac fibrosis, and significant increases in the expression levels of genes (ANP, BNP, and MHCß) associated with cardiac hypertrophy and the chemical staining of the collagen abundance as an indication of fibrosis. In addition, Ang II caused a significant increase in expression levels of inflammatory cytokines (IL-6, IL-17, MIP-2, and IFNγ), NLRP3 inflammasomes, AT1 receptor, and a decrease in AT2 receptor. Treatment with WFA rescued the LV functions and heart hypertrophy and fibrosis. Our results demonstrated, for the first time, that, while WFA has anti-tumorigenic properties, it also ameliorates the cardiac dysfunction induced by Ang II, suggesting that it could be an anticachectic agent that induces direct effects on cardiac muscles.


Sujet(s)
Angiotensine-II , Cachexie , Myocarde , Withanolides , Animaux , Souris , Cachexie/traitement médicamenteux , Cachexie/anatomopathologie , Cardiomégalie/traitement médicamenteux , Cardiomégalie/anatomopathologie , Cytokines/métabolisme , Fibrose , Souris de lignée C57BL , Myocarde/anatomopathologie , Myocarde/métabolisme , Withanolides/pharmacologie , Withanolides/usage thérapeutique
5.
Lipids Health Dis ; 23(1): 144, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38760797

RÉSUMÉ

BACKGROUND: Cancer-associated cachexia (CAC) arises from malignant tumors and leads to a debilitating wasting syndrome. In the pathophysiology of CAC, the depletion of fat plays an important role. The mechanisms of CAC-induced fat loss include the enhancement of lipolysis, inhibition of lipogenesis, and browning of white adipose tissue (WAT). However, few lipid-metabolic enzymes have been reported to be involved in CAC. This study hypothesized that ELOVL6, a critical enzyme for the elongation of fatty acids, may be involved in fat loss in CAC. METHODS: Transcriptome sequencing technology was used to identify CAC-related genes in the WAT of a CAC rodent model. Then, the expression level of ELOVL6 and the fatty acid composition were analyzed in a large clinical sample. Elovl6 was knocked down by siRNA in 3T3-L1 mouse preadipocytes to compare with wild-type 3T3-L1 cells treated with tumor cell conditioned medium. RESULTS: In the WAT of patients with CAC, a significant decrease in the expression of ELOVL6 was found, which was linearly correlated with the extent of body mass reduction. Gas chromatographic analysis revealed an increase in palmitic acid (C16:0) and a decrease in linoleic acid (C18:2n-6) in these tissue samples. After treatment with tumor cell-conditioned medium, 3T3-L1 mouse preadipocytes showed a decrease in Elovl6 expression, and Elovl6-knockdown cells exhibited a reduction in preadipocyte differentiation and lipogenesis. Similarly, the knockdown of Elovl6 in 3T3-L1 cells resulted in a significant increase in palmitic acid (C16:0) and a marked decrease in oleic acid (C18:1n-9) content. CONCLUSION: Overall, the expression of ELOVL6 was decreased in the WAT of CAC patients. Decreased expression of ELOVL6 might induce fat loss in CAC patients by potentially altering the fatty acid composition of adipocytes. These findings suggest that ELOVL6 may be used as a valuable biomarker for the early diagnosis of CAC and may hold promise as a target for future therapies.


Sujet(s)
Cellules 3T3-L1 , Tissu adipeux blanc , Cachexie , Fatty acid elongases , Tumeurs , Fatty acid elongases/génétique , Fatty acid elongases/métabolisme , Animaux , Cachexie/génétique , Cachexie/métabolisme , Cachexie/anatomopathologie , Souris , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/anatomopathologie , Humains , Tumeurs/génétique , Tumeurs/métabolisme , Tumeurs/complications , Tumeurs/anatomopathologie , Mâle , Femelle , Acide palmitique/métabolisme , Lipogenèse/génétique , Adulte d'âge moyen , Acides gras/métabolisme
6.
EMBO Rep ; 25(6): 2592-2609, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38671295

RÉSUMÉ

Various cytokines have been implicated in cancer cachexia. One such cytokine is IL-6, deemed as a key cachectic factor in mice inoculated with colon carcinoma 26 (C26) cells, a widely used cancer cachexia model. Here we tested the causal role of IL-6 in cancer cachexia by knocking out the IL-6 gene in C26 cells. We found that the growth of IL-6 KO tumors was dramatically delayed. More strikingly, while IL-6 KO tumors eventually reached the similar size as wild-type tumors, cachexia still took place, despite no elevation in circulating IL-6. In addition, the knockout of leukemia inhibitory factor (LIF), another IL-6 family cytokine proposed as a cachectic factor in the model, also affected tumor growth but not cachexia. We further showed an increase in the infiltration of immune cell population in the IL-6 KO tumors compared with wild-type controls and the defective IL-6 KO tumor growth was rescued in immunodeficient mice while cachexia was not. Thus, IL-6 promotes tumor growth by facilitating immune evasion but is dispensable for cachexia.


Sujet(s)
Cachexie , Interleukine-6 , Souris knockout , Animaux , Souris , Cachexie/anatomopathologie , Cachexie/génétique , Cachexie/métabolisme , Cachexie/étiologie , Cachexie/immunologie , Lignée cellulaire tumorale , Prolifération cellulaire , Tumeurs du côlon/immunologie , Tumeurs du côlon/génétique , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/métabolisme , Échappement immunitaire , Interleukine-6/métabolisme , Interleukine-6/génétique , Facteur inhibiteur de la leucémie/métabolisme , Facteur inhibiteur de la leucémie/génétique
7.
Cancer Cell ; 42(5): 885-903.e4, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38608702

RÉSUMÉ

With limited treatment options, cachexia remains a major challenge for patients with cancer. Characterizing the interplay between tumor cells and the immune microenvironment may help identify potential therapeutic targets for cancer cachexia. Herein, we investigate the critical role of macrophages in potentiating pancreatic cancer induced muscle wasting via promoting TWEAK (TNF-like weak inducer of apoptosis) secretion from the tumor. Specifically, depletion of macrophages reverses muscle degradation induced by tumor cells. Macrophages induce non-autonomous secretion of TWEAK through CCL5/TRAF6/NF-κB pathway. TWEAK promotes muscle atrophy by activating MuRF1 initiated muscle remodeling. Notably, tumor cells recruit and reprogram macrophages via the CCL2/CCR2 axis and disrupting the interplay between macrophages and tumor cells attenuates muscle wasting. Collectively, this study identifies a feedforward loop between pancreatic cancer cells and macrophages, underlying the non-autonomous activation of TWEAK secretion from tumor cells thereby providing promising therapeutic targets for pancreatic cancer cachexia.


Sujet(s)
Cachexie , Cytokine TWEAK , Macrophages , Tumeurs du pancréas , Cachexie/métabolisme , Cachexie/étiologie , Cachexie/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/complications , Cytokine TWEAK/métabolisme , Animaux , Humains , Macrophages/métabolisme , Souris , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Microenvironnement tumoral , Amyotrophie/métabolisme , Amyotrophie/étiologie , Amyotrophie/anatomopathologie , Chimiokine CCL5/métabolisme , Transduction du signal , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteurs de nécrose tumorale/métabolisme , Récepteurs CCR2/métabolisme , Chimiokine CCL2/métabolisme , Souris de lignée C57BL
8.
Gen Comp Endocrinol ; 353: 114513, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38604437

RÉSUMÉ

Skeletal muscle, comprising a significant proportion (40 to 50 percent) of total body weight in humans, plays a critical role in maintaining normal physiological conditions. Muscle atrophy occurs when the rate of protein degradation exceeds protein synthesis. Sarcopenia refers to age-related muscle atrophy, while cachexia represents a more complex form of muscle wasting associated with various diseases such as cancer, heart failure, and AIDS. Recent research has highlighted the involvement of signaling pathways, including IGF1-Akt-mTOR, MuRF1-MAFbx, and FOXO, in regulating the delicate balance between muscle protein synthesis and breakdown. Myostatin, a member of the TGF-ß superfamily, negatively regulates muscle growth and promotes muscle atrophy by activating Smad2 and Smad3. It also interacts with other signaling pathways in cachexia and sarcopenia. Inhibition of myostatin has emerged as a promising therapeutic approach for sarcopenia and cachexia. Additionally, other TGF-ß family members, such as TGF-ß1, activin A, and GDF11, have been implicated in the regulation of skeletal muscle mass. Furthermore, myostatin cooperates with these family members to impair muscle differentiation and contribute to muscle loss. This review provides an overview of the significance of myostatin and other TGF-ß signaling pathway members in muscular dystrophy, sarcopenia, and cachexia. It also discusses potential novel therapeutic strategies targeting myostatin and TGF-ß signaling for the treatment of muscle atrophy.


Sujet(s)
Cachexie , Amyotrophie , Myostatine , Tumeurs , Sarcopénie , Transduction du signal , Facteur de croissance transformant bêta , Humains , Cachexie/métabolisme , Cachexie/anatomopathologie , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Sarcopénie/métabolisme , Sarcopénie/anatomopathologie , Transduction du signal/physiologie , Tumeurs/métabolisme , Tumeurs/complications , Tumeurs/anatomopathologie , Facteur de croissance transformant bêta/métabolisme , Myostatine/métabolisme , Animaux , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie
9.
Anticancer Res ; 44(5): 1963-1971, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38677769

RÉSUMÉ

BACKGROUND/AIM: Cancer cachexia is a wasting syndrome that has a devastating impact on the prognosis of patients with cancer. It is well-documented that pro-inflammatory cytokines are involved in the progression of this disorder. Therefore, this study was conducted to investigate the protective effect of taurine, an essential nonprotein amino acid with great anti-inflammatory properties, in attenuating muscle atrophy induced by cancer. MATERIALS AND METHODS: Conditioned media (CM) derived from T24 human bladder carcinoma cells with or without 5 mM taurine were incubated with human skeletal muscle cells (HSkMCs) and their differentiation was examined. The intracellular reactive oxygen species (ROS), morphology, and the catabolic pathway were monitored. RESULTS: T24-derived CM with high levels of TNF-α and IL-6 caused aberrant ROS accumulation and formation of atrophic myotubes by HSkMCs. In T24 cancer cells, taurine significantly inhibited the production of TNF-α and IL-6. In HSkMCs, taurine increased ROS clearance during differentiation and preserved the myotube differentiation ability impaired by the inflammatory tumor microenvironment. In addition, taurine ameliorated myotube atrophy by regulating the Akt/FoxO1/MuRF1 and MAFbx signaling pathways. CONCLUSION: Taurine rescues cancer-induced atrophy in human skeletal muscle cells by ameliorating the inflammatory tumor microenvironment. Taurine supplementation may be a promising approach for intervening with the progression of cancer cachexia.


Sujet(s)
Amyotrophie , Espèces réactives de l'oxygène , Taurine , Microenvironnement tumoral , Humains , Taurine/pharmacologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Amyotrophie/anatomopathologie , Amyotrophie/traitement médicamenteux , Amyotrophie/métabolisme , Amyotrophie/étiologie , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/anatomopathologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Cachexie/traitement médicamenteux , Cachexie/anatomopathologie , Cachexie/métabolisme , Cachexie/étiologie , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/métabolisme , Milieux de culture conditionnés/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/métabolisme
10.
Am J Physiol Cell Physiol ; 326(5): C1520-C1542, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38557354

RÉSUMÉ

Cancer cachexia is the result of complex interorgan interactions initiated by cancer cells and changes in patient behavior such as decreased physical activity and energy intake. Therefore, it is crucial to distinguish between the direct and indirect effects of cancer cells on muscle mass regulation and bioenergetics to identify novel therapeutic targets. In this study, we investigated the direct effects of Colon-26 cancer cells on the molecular regulating machinery of muscle mass and its bioenergetics using a coculture system with C2C12 myotubes. Our results demonstrated that coculture with Colon-26 cells induced myotube atrophy and reduced skeletal muscle protein synthesis and its regulating mechanistic target of rapamycin complex 1 signal transduction. However, we did not observe any activating effects on protein degradation pathways including ubiquitin-proteasome and autophagy-lysosome systems. From a bioenergetic perspective, coculture with Colon-26 cells decreased the complex I-driven, but not complex II-driven, mitochondrial ATP production capacity, while increasing glycolytic enzyme activity and glycolytic metabolites, suggesting a shift in energy metabolism toward glycolysis dominance. Gene expression profiling by RNA sequencing showed that the increased activity of glycolytic enzymes was consistent with changes in gene expression. However, the decreased ATP production capacity of mitochondria was not in line with the gene expression. The potential direct interaction between cancer cells and skeletal muscle cells revealed in this study may contribute to a better fundamental understanding of the complex pathophysiology of cancer cachexia.NEW & NOTEWORTHY We explored the potential direct interplay between colon cancer cells (Colon-26) and skeletal muscle cells (C2C12 myotubes) employing a noncontact coculture experimental model. Our findings reveal that coculturing with Colon-26 cells substantially impairs the protein synthesis rate, concurrently instigating a metabolic shift toward glycolytic dominance in C2C12 myotubes. This research unveils critical insights into the intricate cellular cross talk underpinning the complex pathophysiology of cancer cachexia.


Sujet(s)
Cachexie , Techniques de coculture , Tumeurs du côlon , Métabolisme énergétique , Glycolyse , Fibres musculaires squelettiques , Fibres musculaires squelettiques/métabolisme , Animaux , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Souris , Lignée cellulaire tumorale , Cachexie/métabolisme , Cachexie/anatomopathologie , Biosynthèse des protéines , Humains , Transduction du signal , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Protéines du muscle/biosynthèse
11.
Mol Brain ; 17(1): 20, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38685046

RÉSUMÉ

While the excessive inflammation in cancer cachexia is well-known to be induced by the overproduction of inflammatory mediators in the periphery, microflora disruption and brain dysfunction are also considered to contribute to the induction of cancer cachexia. Hypothalamic microglia play a crucial role in brain inflammation and central-peripheral immune circuits via the production of inflammatory mediators. In the present study, we evaluated possible changes in excessive secretion of gut microbiota-derived endotoxin and the expression timeline of several inflammation-regulatory mediators and their inhibiting modulators in hypothalamic microglia of a mouse model of cancer cachexia following transplantation of pancreatic cancer cells. We demonstrated that the plasma level of lipopolysaccharide (LPS) was significantly increased with an increase in anaerobic bacteria, especially Firmicutes, in the gut at the late stage of tumor-bearing mice that exhibited dramatic appetite loss, sarcopenia and severe peripheral immune suppression. At the early stage, in which tumor-bearing mice had not yet displayed "cachexia symptoms", the mRNA expression of pro-inflammatory cytokines, but not of the neurodegenerative and severe inflammatory modulator lipocalin-2 (LCN2), was significantly increased, whereas at the late "cachexia stage", the level of LCN2 mRNA was significantly increased along with significant decreases in levels of inhibitory immune checkpoint receptors programmed death receptor-1 (PD-1) and CD112R in hypothalamic microglia. In addition, a high density of activated neurons in the paraventricular nucleus (PVN) of the hypothalamus region and a significant increase in corticosterone secretion were found in cachexia model mice. Related to the cachexia state, released corticosterone was clearly increased in normal mice with specific activation of PVN neurons. A marked decrease in the natural killer cell population was also observed in the spleen of mice with robust activation of PVN neurons as well as mice with cancer cachexia. On the other hand, in vivo administration of LPS in normal mice induced hypothalamic microglia with low expression of inhibitory immune checkpoint receptors. These findings suggest that the induction of cancer cachexia may parallel exacerbation of the hypothalamic inflammatory status with polarization to microglia expressed with low levels of inhibitory immune checkpoint receptors following LPS release from the gut microflora.


Sujet(s)
Cachexie , Hypothalamus , Lipocaline-2 , Lipopolysaccharides , Microglie , Animaux , Cachexie/complications , Cachexie/anatomopathologie , Microglie/métabolisme , Hypothalamus/métabolisme , Lipocaline-2/métabolisme , Lipopolysaccharides/pharmacologie , Mâle , Lignée cellulaire tumorale , Souris , Récepteur-1 de mort cellulaire programmée/métabolisme , Microbiome gastro-intestinal , Cytokines/métabolisme , Tumeurs/complications , Souris de lignée C57BL , Médiateurs de l'inflammation/métabolisme , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique
12.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189103, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38679401

RÉSUMÉ

Cancer-associated cachexia (CAC) is a complex multiple organ syndrome that significantly contributes to reduced quality of life and increased mortality among many cancer patients. Its multifactorial nature makes its early diagnosis and effective therapeutic interventions challenging. Adipose tissue is particularly impacted by cachexia, typically through increased lipolysis, browning and thermogenesis, mainly at the onset of the disease. These processes lead to depletion of fat mass and contribute to the dysfunction of other organs. The ß-adrenergic signalling pathways are classical players in the regulation of adipose tissue metabolism. They are activated upon sympathetic stimulation inducing lipolysis, browning and thermogenesis, therefore contributing to energy expenditure. Despite accumulating evidence suggesting that ß3-adrenergic receptor stimulation may be crucial to the adipose tissue remodelling during cachexia, the literature remains controversial. Moreover, there is limited knowledge regarding sexual dimorphism of adipose tissue in the context of cachexia. This review paper aims to present the current knowledge regarding adipose tissue wasting during CAC, with a specific focus on the role of the ß3-adrenergic receptor, placing it as a potential therapeutic target against cachexia.


Sujet(s)
Tissu adipeux , Cachexie , Lipolyse , Tumeurs , Récepteurs bêta-3 adrénergiques , Transduction du signal , Cachexie/métabolisme , Cachexie/anatomopathologie , Cachexie/étiologie , Humains , Tumeurs/complications , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Récepteurs bêta-3 adrénergiques/métabolisme , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Métabolisme énergétique , Thermogenèse , Animaux
13.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167179, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38653357

RÉSUMÉ

Muscle degeneration is a common feature in cancer cachexia that cannot be reversed. Recent advances show that the endocannabinoid system, and more particularly cannabinoid receptor 1 (CB1), regulates muscle processes, including metabolism, anabolism and regenerative capacity. However, it is unclear whether muscle endocannabinoids, their receptors and enzymes are responsive to cachexia and exercise. Therefore, this study investigated whether cachexia and exercise affected muscle endocannabinoid signaling, and whether CB1 expression correlated with markers of muscle anabolism, catabolism and metabolism. Male BALB/c mice were injected with PBS (CON) or C26 colon carcinoma cells (C26) and had access to wheel running (VWR) or remained sedentary (n = 5-6/group). Mice were sacrificed 18 days upon PBS/tumor cell injection. Cachexic mice exhibited a lower muscle CB1 expression (-43 %; p < 0.001) and lower levels of the endocannabinoid anandamide (AEA; -22 %; p = 0.044), as well as a lower expression of the AEA-synthesizing enzyme NAPE-PLD (-37 %; p < 0.001), whereas the expression of the AEA degrading enzyme FAAH was higher (+160 %; p < 0.001). The 2-AG-degrading enzyme MAGL, was lower in cachexic muscle (-34 %; p = 0.007), but 2-AG and its synthetizing enzyme DAGLß were not different between CON and C26. VWR increased muscle CB1 (+25 %; p = 0.005) and increased MAGL expression (+30 %; p = 0.035). CB1 expression correlated with muscle mass, markers of metabolism (e.g. p-AMPK, PGC1α) and of catabolism (e.g. p-FOXO, LC3b, Atg5). Our findings depict an emerging role of the endocannabinoid system in muscle physiology. Future studies should elaborate how this translates into potential therapies to combat cancer cachexia, and other degenerative conditions.


Sujet(s)
Cachexie , Endocannabinoïdes , Souris de lignée BALB C , Muscles squelettiques , Récepteur cannabinoïde de type CB1 , Animaux , Endocannabinoïdes/métabolisme , Mâle , Souris , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Cachexie/métabolisme , Cachexie/anatomopathologie , Récepteur cannabinoïde de type CB1/métabolisme , Récepteur cannabinoïde de type CB1/génétique , Lignée cellulaire tumorale , Amides gras polyinsaturés N-alkylés/métabolisme , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Conditionnement physique d'animal , Acides arachidoniques/métabolisme
14.
J Transl Med ; 22(1): 408, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38689293

RÉSUMÉ

Cancer-related cachexia is a metabolic syndrome characterized by weight loss, adipose tissue decomposition, and progressive skeletal muscle atrophy. It is a major complication of many advanced cancers and seriously affects the quality of life and survival of cancer patients. However, the specific molecules that mediate cancer-related cachexia remain elusive, and the fundamental cellular and molecular mechanisms associated with muscle atrophy and lipidolysis in cancer patients still need to be investigated. Exosomes, a newly discovered class of small extracellular vesicles that facilitate intercellular communication, have a significant role in the onset and development of various cancers. Studies have shown that exosomes play a role in the onset and progression of cancer-related cachexia by transporting active molecules such as nucleic acids and proteins. This review aimed to provide an overview of exosome developments in cancer-induced skeletal muscle atrophy and adipose tissue degradation. More importantly, exosomes were shown to have potential as diagnostic markers or therapeutic strategies for cachexia and were prospected, providing novel strategies for the diagnosis and treatment of cancer-related cachexia.


Sujet(s)
Cachexie , Exosomes , Tumeurs , Cachexie/étiologie , Cachexie/anatomopathologie , Cachexie/thérapie , Cachexie/métabolisme , Humains , Exosomes/métabolisme , Tumeurs/complications , Tumeurs/anatomopathologie , Animaux , Tissu adipeux/anatomopathologie , Tissu adipeux/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/métabolisme , Amyotrophie/étiologie
15.
Eur J Pharmacol ; 974: 176538, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38552940

RÉSUMÉ

Chemotherapy is one of the primary and indispensable intervention against cancers though it is always accompanied by severe side effects especially cachexia. Cachexia is a fatal metabolic disorder syndrome, mainly characterized by muscle loss. Oxidative stress is the key factor that trigger cachectic muscle loss by inducing imbalance in protein metabolism and apoptosis. Here, we showed an oral compound (Z526) exhibited potent alleviating effects on C2C12 myotube atrophy induced by various chemotherapeutic agents in vitro as well as mice muscle loss and impaired grip force induced by oxaliplatin in vivo. Furthermore, Z526 also could ameliorate C2C12 myotube atrophy induced by the combination of chemotherapeutic agents with conditioned medium of various tumor cells in vitro as well as mice muscle atrophy of C26 tumor-bearing mice treated with oxaliplatin. The pharmacological effects of Z526 were based on its potency in reducing oxidative stress in cachectic myocytes and muscle tissues, which inhibited the activation of NF-κB and STAT3 to decrease Atrogin-1-mediated protein degradation, activated the AKT/mTOR signaling pathway to promote protein synthesis, regulated Bcl-2/BAX ratio to reduce Caspase-3-triggered apoptosis. Our work suggested Z526 to be an optional strategy for ameliorating cachexia muscle atrophy in the multimodality treatment of cancers.


Sujet(s)
Antinéoplasiques , Apoptose , Cachexie , Amyotrophie , Stress oxydatif , Animaux , Cachexie/traitement médicamenteux , Cachexie/anatomopathologie , Cachexie/induit chimiquement , Cachexie/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/effets indésirables , Amyotrophie/traitement médicamenteux , Amyotrophie/induit chimiquement , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/anatomopathologie , Lignée cellulaire tumorale , Facteur de transcription STAT-3/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Souris de lignée BALB C , Lignée cellulaire , Protéines du muscle/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie
16.
Nutrition ; 121: 112365, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38377700

RÉSUMÉ

OBJECTIVES: The practicality and effectiveness of using the prognostic value of the neutrophil-to-albumin ratio (NAR) in evaluating patients with cancer remain unclear, and research is needed to fully understand its potential application in the cancer population. METHODS: The Kaplan-Meier method was used for survival analysis, and the log-rank test was employed for comparison. Univariate and multivariate Cox proportional hazards models were used to determine the prognostic biomarkers, and Logistic regression analysis was conducted to investigate the relationship between NAR and 90-day outcomes and cachexia. RESULTS: The study included 14 682 patients with cancer, divided into discovery (6592 patients), internal validation (2820 patients), and external validation groups (5270 patients). Patients with high NAR had higher all-cause mortality than those with low NAR in the discovery (50.15% versus 69.29%, P < 0.001), internal validation (54.18% versus 70.91%, P < 0.001), and external validation cohorts (40.60% versus 66.68%, P < 0.001). In the discovery cohort, high NAR was observed to be independently associated with all-cause mortality in patients (HR 1.16, 95% CI 1.12-1.19; P < 0.001). Moreover, we validated the promising prognostic value of NAR as a predictor of survival in patients with cancer through internal validation (HR 1.21, 95% CI 1.16-1.27, P < 0.001) and external validation cohorts (HR 1.27, 95% CI 1.21-1.34, P < 0.001). Additionally, in the subgroup analysis by tumor type, high NAR was identified as a risk factor for most cancers, except for breast cancer. CONCLUSIONS: This study showed that NAR is a feasible and promising biomarker for predicting prognosis and cancer cachexia in cancer patients.


Sujet(s)
Tumeurs , Granulocytes neutrophiles , Humains , Pronostic , Cachexie/anatomopathologie , Tumeurs/complications , Tumeurs/anatomopathologie , Albumines , Études de cohortes , Études rétrospectives
17.
J Cachexia Sarcopenia Muscle ; 15(1): 255-269, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38062876

RÉSUMÉ

BACKGROUND: Skeletal muscle atrophy, particularly ageing-related muscular atrophy such as sarcopenia, is a significant health concern. Despite its prevalence, the underlying mechanisms remain poorly understood, and specific approved medications are currently unavailable. Deleted in breast cancer 1 (DBC1) is a well-known regulator of senescence, metabolism or apoptosis. Recent reports suggest that DBC1 may also potentially regulate muscle function, as mice lacking DBC1 exhibit weakness and limpness. However, the function of DBC1 in skeletal muscle and its associated molecular mechanisms remain unknown, thus prompting the focus of this study. METHODS: Tibialis anterior (TA) muscle-specific DBC1 knockdown C57BL/6J male mice were generated through a single injection of 2.00 E + 11 vg of adeno-associated virus 9 delivering single-guide RNA for DBC1. Grip strength and endurance were assessed 2 months later, followed by skeletal muscle harvest. Muscle atrophy model was generated by cast immobilization of the mouse hindlimb for 2 weeks. Molecular markers of atrophy were probed in muscles upon termination. Cardiotoxin (CTX) was injected in TA muscles of DBC1 knockdown mice, and muscle regeneration was assessed by immunohistochemistry, quantitative PCR and western blotting. DBC1 knockdown C2C12 cells and myotubes were investigated using immunofluorescence staining, Seahorse, immunohistology, fluorescence-activated cell sorting and RNA-sequencing analyses. RESULTS: DBC1 knockdown in skeletal muscle of young mice led to signatures of muscle atrophy, including a 28% reduction in muscle grip force (P = 0.023), a 54.4% reduction in running distance (P = 0.002), a 14.3% reduction in muscle mass (P = 0.007) and significantly smaller myofibre cross-sectional areas (P < 0.0001). DBC1 levels decrease in age-related or limb immobilization-induced atrophic mouse muscles and overexpress DBC1-attenuated atrophic phenotypes in these mice. Muscle regeneration was hampered in mice with CTX-induced muscle injury by DBC1 knockdown, as evidenced by reductions in myofibre cross-sectional areas of regenerating myofibres with centralized nuclei (P < 0.0001), percentages of MyoG+ nuclei (P < 0.0001) and fusion index (P < 0.0001). DBC1 transcriptionally regulated mouse double minute 2 (MDM2), which mediated ubiquitination and degradation of forkhead box O3 (FOXO3). Increased FOXO3 proteins hampered myogenesis in DBC1 knockdown satellite cells by compromising around 50% of mitochondrial functions (P < 0.001) and exacerbated atrophy in DBC1 knockdown myofibres by activating the ubiquitin-proteasome and autophagy-lysosome pathways. CONCLUSIONS: DBC1 is essential in maintaining skeletal muscle integrity by protecting against myofibres wasting and enhancing muscle regeneration via FOXO3. This research highlights the significance of DBC1 for healthy skeletal muscle function and its connection to muscular atrophy.


Sujet(s)
Muscles squelettiques , , Animaux , Mâle , Souris , Cachexie/anatomopathologie , Souris de lignée C57BL , Développement musculaire , Muscles squelettiques/anatomopathologie , Amyotrophie/anatomopathologie
18.
J Cachexia Sarcopenia Muscle ; 15(1): 124-137, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38062911

RÉSUMÉ

BACKGROUND: More than 650 million people are obese (BMI > 30) worldwide, which increases their risk for several metabolic diseases and cancer. While cachexia and obesity are at opposite ends of the weight spectrum, leading many to suggest a protective effect of obesity against cachexia, mechanistic support for obesity's benefit is lacking. Given that obesity and cachexia are both accompanied by metabolic dysregulation, we sought to investigate the impact of obesity on skeletal muscle mass loss and mitochondrial dysfunction in murine cancer cachexia. METHODS: Male C57BL/6 mice were given a purified high fat or standard diet for 16 weeks before being implanted with 106 Lewis lung carcinoma (LLC) cells. Mice were monitored for 25 days, and hindlimb muscles were collected for cachexia indices and mitochondrial assessment via western blotting, high-resolution respirometry and transmission electron microscopy (TEM). RESULTS: Obese LLC mice experienced significant tumour-free body weight loss similar to lean (-12.8% vs. -11.8%, P = 0.0001) but had reduced survival (33.3% vs. 6.67%, χ2  = 10.04, P = 0.0182). Obese LLC mice had reduced muscle weights (-24%, P < 0.0354) and mCSA (-16%, P = 0.0004) with similar activation of muscle p65 (P = 0.0337), and p38 (P = 0.0008). ADP-dependent coupled respiration was reduced in both Obese and Obese LLC muscle (-30%, P = 0.0072) consistent with reductions in volitional cage activity (-39%, P < 0.0001) and grip strength (-41%, P < 0.0001). TEM revealed stepwise reductions in intermyofibrillar and subsarcolemmal mitochondrial size with Obese (IMF: -37%, P = 0.0009; SS: -21%, P = 0.0101) and LLC (IMF: -40%, P = 0.0019; SS: -27%, P = 0.0383) mice. Obese LLC mice had increased pAMPK (T172; P = 0.0103) and reduced FIS1 (P = 0.0029) and DRP1 (P < 0.0001) mitochondrial fission proteins, which were each unchanged in Lean LLC. Further, mitochondrial TEM analysis revealed that Obese LLC mice had an accumulation of damaged and dysfunctional mitochondria (IMF: 357%, P = 0.0395; SS: 138%, P = 0.0174) in concert with an accumulation of p62 (P = 0.0328) suggesting impaired autophagy and clearance of damaged mitochondria. Moreover, we observed increases in electron lucent vacuoles only in Obese LLC muscle (IMF: 421%, P = 0.0260; SS: 392%, P = 0.0192), further supporting an accumulation of damaged materials that cannot be properly cleared in the obese cachectic muscle. CONCLUSIONS: Taken together, these results demonstrate that obesity is not protective against cachexia and suggest exacerbated impairments to mitochondrial function and quality control with a particular disruption in the removal of damaged mitochondria. Our findings highlight the need for consideration of the severity of obesity and pre-existing metabolic conditions when determining the impact of weight status on cancer-induced cachexia and functional mitochondrial deficits.


Sujet(s)
Cachexie , Carcinome pulmonaire de Lewis , Humains , Mâle , Animaux , Souris , Cachexie/anatomopathologie , Souris de lignée C57BL , Mitochondries/métabolisme , Amyotrophie/anatomopathologie , Carcinome pulmonaire de Lewis/complications , Carcinome pulmonaire de Lewis/anatomopathologie , Obésité/complications , Obésité/anatomopathologie , Muscles squelettiques/anatomopathologie
19.
Chin Med J (Engl) ; 137(1): 44-62, 2024 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-37968131

RÉSUMÉ

ABSTRACT: Cancer cachexia is a multi-organ syndrome and closely related to changes in signal communication between organs, which is mediated by cancer cachexia factors. Cancer cachexia factors, being the general name of inflammatory factors, circulating proteins, metabolites, and microRNA secreted by tumor or host cells, play a role in secretory or other organs and mediate complex signal communication between organs during cancer cachexia. Cancer cachexia factors are also a potential target for the diagnosis and treatment. The pathogenesis of cachexia is unclear and no clear effective treatment is available. Thus, the treatment of cancer cachexia from the perspective of the tumor ecosystem rather than from the perspective of a single molecule and a single organ is urgently needed. From the point of signal communication between organs mediated by cancer cachexia factors, finding a deeper understanding of the pathogenesis, diagnosis, and treatment of cancer cachexia is of great significance to improve the level of diagnosis and treatment. This review begins with cancer cachexia factors released during the interaction between tumor and host cells, and provides a comprehensive summary of the pathogenesis, diagnosis, and treatment for cancer cachexia, along with a particular sight on multi-organ signal communication mediated by cancer cachexia factors. This summary aims to deepen medical community's understanding of cancer cachexia and may conduce to the discovery of new diagnostic and therapeutic targets for cancer cachexia.


Sujet(s)
Cachexie , Tumeurs , Humains , Cachexie/étiologie , Cachexie/métabolisme , Cachexie/anatomopathologie , Écosystème , Tumeurs/métabolisme , Syndrome , Muscles squelettiques/anatomopathologie
20.
Am J Clin Oncol ; 47(2): 49-55, 2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38011024

RÉSUMÉ

BACKGROUND: Cancer cachexia is a syndrome of unintentional weight loss resulting in progressive functional impairment. Knowledge of radiation therapy utilization in patients with cancer cachexia is limited. We evaluated the use of curative and palliative-intent radiation for the management of patients with non-small cell lung cancer (NSCLC) with cachexia to determine whether tumor-directed therapy affected cachexia-associated outcomes. METHODS: Using an Institutional Tumor Registry, we evaluated all patients with stages of NSCLC treated at a tertiary care system from 2006 to 2013. We adopted the international consensus definition for cachexia, with staging designated by the registry and positron emission tomography. Radiotherapy delivery and intent were retrospectively assessed. RESULTS: In total, 1330 patients with NSCLC were analyzed. Curative-intent radiotherapy was utilized equally between patients with cachexia and non-cachexia with stages I to III NSCLC. Conversely, significantly more patients with stage IV disease and cachexia received palliative radiotherapy versus those without (74% vs 63%, P = 0.006). Cachexia-associated survival was unchanged irrespective of tumor-directed radiation therapy with curative or palliative intent. In fact, pretreatment cachexia was associated with reduced survival for patients with stage III NSCLC receiving curative-intent radiotherapy (median survival = 23.9 vs 15.0 mo, P = 0.009). Finally, multivariate analysis identified pretreatment cachexia as an independent variable associated with worsened survival (hazard ratio = 1.31, CI: 1.14,1.52). CONCLUSION: Patients with advanced NSCLC with cachexia received more palliative-intent radiation than those without weight loss. Tumor-directed therapy in either a curative or palliative approach failed to alter cachexia patient survival across all stages of the disease. These findings offer critical information on the appropriate utilization of radiation in the management of patients with NSCLC with cachexia.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Carcinome pulmonaire non à petites cellules/complications , Carcinome pulmonaire non à petites cellules/radiothérapie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Tumeurs du poumon/complications , Tumeurs du poumon/radiothérapie , Tumeurs du poumon/traitement médicamenteux , Cachexie/étiologie , Cachexie/anatomopathologie , Études rétrospectives , Stadification tumorale , Perte de poids
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...