Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
1.
Physiol Rep ; 12(18): e70044, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39294861

RESUMEN

Cancer cachexia manifests as whole body wasting, however, the precise mechanisms governing the alterations in skeletal muscle and cardiac anabolism have yet to be fully elucidated. In this study, we explored changes in anabolic processes in both skeletal and cardiac muscles in the Yoshida AH-130 ascites hepatoma model of cancer cachexia. AH-130 tumor-bearing rats experienced significant losses in body weight, skeletal muscle, and heart mass. Skeletal and cardiac muscle loss was associated with decreased ribosomal (r)RNA, and hypophosphorylation of the eukaryotic factor 4E binding protein 1. Endoplasmic reticulum stress was evident by higher activating transcription factor mRNA in skeletal muscle and growth arrest and DNA damage-inducible protein (GADD)34 mRNA in both skeletal and cardiac muscles. Tumors provoked an increase in tissue expression of interferon-γ in the heart, while an increase in interleukin-1ß mRNA was apparent in both skeletal and cardiac muscles. We conclude that compromised skeletal muscle and heart mass in the Yoshida AH-130 ascites hepatoma model involves a marked reduction translational capacity and efficiency. Furthermore, our observations suggest that endoplasmic reticulum stress and tissue production of pro-inflammatory factors may play a role in the development of skeletal and cardiac muscle wasting.


Asunto(s)
Caquexia , Músculo Esquelético , Miocardio , Respuesta de Proteína Desplegada , Animales , Caquexia/metabolismo , Caquexia/etiología , Caquexia/patología , Caquexia/genética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratas , Miocardio/metabolismo , Miocardio/patología , Ratas Wistar , Estrés del Retículo Endoplásmico , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética
2.
J Physiol ; 602(17): 4215-4235, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39167700

RESUMEN

Oxidative stress contributes to the loss of skeletal muscle mass and function in cancer cachexia. However, this outcome may be mitigated by an improved endogenous antioxidant defence system. Here, using the well-established oxidative stress-inducing muscle atrophy model of Lewis lung carcinoma (LLC) in 13-week-old male C57BL/6J mice, we demonstrate that extracellular superoxide dismutase (EcSOD) levels increase in the cachexia-prone extensor digitorum longus muscle. LLC transplantation significantly increased interleukin-1ß (IL-1ß) expression and release from extensor digitorum longus muscle fibres. Moreover, IL-1ß treatment of C2C12 myotubes increased NBR1, p62 phosphorylation at Ser351, Nrf2 nuclear translocation and EcSOD protein expression. Additional studies in vivo indicated that intramuscular IL-1ß injection is sufficient to stimulate EcSOD expression, which is prevented by muscle-specific knockout of p62 and Nrf2 (i.e. in p62 skmKO and Nrf2 skmKO mice, respectively). Finally, since an increase in circulating IL-1ß may lead to unwanted outcomes, we demonstrate that targeting this pathway at p62 is sufficient to drive muscle EcSOD expression in an Nrf2-dependent manner. In summary, cancer cachexia increases EcSOD expression in extensor digitorum longus muscle via muscle-derived IL-1ß-induced upregulation of p62 phosphorylation and Nrf2 activation. These findings provide further mechanistic evidence for the therapeutic potential of p62 and Nrf2 to mitigate cancer cachexia-induced muscle atrophy. KEY POINTS: Oxidative stress plays an important role in muscle atrophy during cancer cachexia. EcSOD, which mitigates muscle loss during oxidative stress, is upregulated in 13-week-old male C57BL/6J mice of extensor digitorum longus muscles during cancer cachexia. Using mouse and cellular models, we demonstrate that cancer cachexia promotes muscle EcSOD protein expression via muscle-derived IL-1ß-dependent stimulation of the NBR1-p62-Nrf2 signalling pathway. These results provide further evidence for the potential therapeutic targeting of the NBR1-p62-Nrf2 signalling pathway downstream of IL-1ß to mitigate cancer cachexia-induced muscle atrophy.


Asunto(s)
Caquexia , Interleucina-1beta , Ratones Endogámicos C57BL , Músculo Esquelético , Factor 2 Relacionado con NF-E2 , Transducción de Señal , Superóxido Dismutasa , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Caquexia/metabolismo , Caquexia/etiología , Caquexia/genética , Masculino , Interleucina-1beta/metabolismo , Músculo Esquelético/metabolismo , Ratones , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/genética , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/complicaciones , Carcinoma Pulmonar de Lewis/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/genética , Ratones Noqueados , Estrés Oxidativo
3.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000167

RESUMEN

Skeletal muscle aging and sarcopenia result in similar changes in the levels of aging markers. However, few studies have examined cancer sarcopenia from the perspective of aging. Therefore, this study investigated aging in cancer sarcopenia and explored its causes in vitro and in vivo. In mouse aging, in vitro cachexia, and mouse cachexia models, skeletal muscles showed similar changes in aging markers including oxidative stress, fibrosis, reduced muscle differentiation potential, and telomere shortening. Furthermore, examination of mitochondrial DNA from skeletal muscle revealed a 5 kb deletion in the major arc; truncation of complexes I, IV, and V in the electron transport chain; and reduced oxidative phosphorylation (OXPHOS). The mouse cachexia model demonstrated high levels of high-mobility group box-1 (HMGB1) and tumor necrosis factor-α (TNFα) in cancer ascites. Continuous administration of neutralizing antibodies against HMGB1 and TNFα in this model reduced oxidative stress and abrogated mitochondrial DNA deletion. These results suggest that in cancer sarcopenia, mitochondrial oxidative stress caused by inflammatory cytokines leads to mitochondrial DNA damage, which in turn leads to decreased OXPHOS and the promotion of aging.


Asunto(s)
Envejecimiento , Daño del ADN , ADN Mitocondrial , Proteína HMGB1 , Músculo Esquelético , Estrés Oxidativo , Sarcopenia , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratones , Envejecimiento/metabolismo , Envejecimiento/genética , Sarcopenia/metabolismo , Sarcopenia/patología , Sarcopenia/genética , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Caquexia/metabolismo , Caquexia/patología , Caquexia/genética , Caquexia/etiología , Fosforilación Oxidativa , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Masculino , Ratones Endogámicos C57BL
4.
Curr Opin Support Palliat Care ; 18(3): 120-125, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39007915

RESUMEN

PURPOSE OF THE REVIEW: Cancer-associated cachexia is a wasting syndrome entailing loss in body mass and a shortened life expectancy. There is currently no effective treatment to abrogate this syndrome, which leads to 20-30% of deaths in patients with cancer. While there have been advancements in defining signaling factors/pathways in cancer-induced muscle wasting, targeting the same in the clinic has not been as successful. Krüppel-like factor 10 (KLF10), a transcription factor implicated in muscle regulation, is regulated by the transforming growth factor-beta signaling pathway. This review proposes KLF10 as a potential convergence point of diverse signaling pathways involved in muscle wasting. RECENT FINDINGS: KLF10 was discovered as a target of transforming growth factor-beta decades ago but more recently it has been shown that deletion of KLF10 rescues cancer-induced muscle wasting. Moreover, KLF10 has also been shown to bind key atrophy genes associated with muscle atrophy in vitro . SUMMARY: There is an elevated need to explore targets in cachexia, which will successfully translate into the clinic. Investigating a convergence point downstream of multiple signaling pathways might hold promise in developing effective therapies for cachexia.


Asunto(s)
Caquexia , Factores de Transcripción de la Respuesta de Crecimiento Precoz , Factores de Transcripción de Tipo Kruppel , Neoplasias , Transducción de Señal , Caquexia/etiología , Caquexia/genética , Caquexia/fisiopatología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias/complicaciones , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Atrofia Muscular/genética , Factor de Crecimiento Transformador beta/metabolismo , Músculo Esquelético/metabolismo
5.
Lipids Health Dis ; 23(1): 144, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760797

RESUMEN

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.


Asunto(s)
Células 3T3-L1 , Tejido Adiposo Blanco , Caquexia , Elongasas de Ácidos Grasos , Neoplasias , Elongasas de Ácidos Grasos/genética , Elongasas de Ácidos Grasos/metabolismo , Animales , Caquexia/genética , Caquexia/metabolismo , Caquexia/patología , Ratones , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/complicaciones , Neoplasias/patología , Masculino , Femenino , Ácido Palmítico/metabolismo , Lipogénesis/genética , Persona de Mediana Edad , Ácidos Grasos/metabolismo
6.
EMBO Rep ; 25(6): 2592-2609, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38671295

RESUMEN

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.


Asunto(s)
Caquexia , Interleucina-6 , Ratones Noqueados , Animales , Ratones , Caquexia/patología , Caquexia/genética , Caquexia/metabolismo , Caquexia/etiología , Caquexia/inmunología , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/metabolismo , Evasión Inmune , Interleucina-6/metabolismo , Interleucina-6/genética , Factor Inhibidor de Leucemia/metabolismo , Factor Inhibidor de Leucemia/genética
7.
BMC Cancer ; 24(1): 414, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38570770

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) presents with a high mortality rate. Two important features of PDAC contribute to this poor outcome. The first is metastasis which occurs in ~ 80% of PDAC patients. The second is cachexia, which compromises treatment tolerance for patients and reduces their quality of life. Although various mouse models of PDAC exist, recapitulating both metastatic and cachectic features have been challenging. METHODS: Here, we optimize an orthotopic mouse model of PDAC by altering several conditions, including the subcloning of parental murine PDAC cells, implantation site, number of transplanted cells, and age of recipient mice. We perform spatial profiling to compare primary and metastatic immune microenvironments and RNA sequencing to gain insight into the mechanisms of muscle wasting in PDAC-induced cachexia, comparing non-metastatic to metastatic conditions. RESULTS: These modifications extend the time course of the disease and concurrently increase the rate of metastasis to approximately 70%. Furthermore, reliable cachexia endpoints are achieved in both PDAC mice with and without metastases, which is reminiscent of patients. We also find that cachectic muscles from PDAC mice with metastasis exhibit a similar transcriptional profile to muscles derived from mice and patients without metastasis. CONCLUSION: Together, this model is likely to be advantageous in both advancing our understanding of the mechanism of PDAC cachexia, as well as in the evaluation of novel therapeutics.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Caquexia/genética , Calidad de Vida , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Fenotipo , Microambiente Tumoral
8.
Comput Biol Med ; 172: 108233, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38452471

RESUMEN

BACKGROUND: Cancer cachexia is a severe metabolic syndrome marked by skeletal muscle atrophy. A successful clinical intervention for cancer cachexia is currently lacking. The study of cachexia mechanisms is largely based on preclinical animal models and the availability of high-throughput transcriptomic datasets of cachectic mouse muscles is increasing through the extensive use of next generation sequencing technologies. METHODS: Cachectic mouse muscle transcriptomic datasets of ten different studies were combined and mined by seven attribute weighting models, which analysed both categorical variables and numerical variables. The transcriptomic signature of cancer cachexia was identified by attribute weighting algorithms and was used to evaluate the performance of eleven pattern discovery models. The signature was employed to find the best combination of drugs (drug repurposing) for developing cancer cachexia treatment strategies, as well as to evaluate currently used cachexia drugs by literature mining. RESULTS: Attribute weighting algorithms ranked 26 genes as the transcriptomic signature of muscle from mice with cancer cachexia. Deep Learning and Random Forest models performed better in differentiating cancer cachexia cases based on muscle transcriptomic data. Literature mining revealed that a combination of melatonin and infliximab has negative interactions with 2 key genes (Rorc and Fbxo32) upregulated in the transcriptomic signature of cancer cachexia in muscle. CONCLUSIONS: The integration of machine learning, meta-analysis and literature mining was found to be an efficient approach to identifying a robust transcriptomic signature for cancer cachexia, with implications for improving clinical diagnosis and management of this condition.


Asunto(s)
Caquexia , Neoplasias , Animales , Ratones , Caquexia/genética , Caquexia/metabolismo , Minería de Datos , Perfilación de la Expresión Génica , Aprendizaje Automático , Metaanálisis como Asunto , Músculo Esquelético , Neoplasias/complicaciones , Neoplasias/genética , Neoplasias/metabolismo
9.
Nat Commun ; 15(1): 627, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38245529

RESUMEN

Cancer cachexia is a systemic metabolic syndrome characterized by involuntary weight loss, and muscle and adipose tissue wasting. Mechanisms underlying cachexia remain poorly understood. Leukemia inhibitory factor (LIF), a multi-functional cytokine, has been suggested as a cachexia-inducing factor. In a transgenic mouse model with conditional LIF expression, systemic elevation of LIF induces cachexia. LIF overexpression decreases de novo lipogenesis and disrupts lipid homeostasis in the liver. Liver-specific LIF receptor knockout attenuates LIF-induced cachexia, suggesting that LIF-induced functional changes in the liver contribute to cachexia. Mechanistically, LIF overexpression activates STAT3 to downregulate PPARα, a master regulator of lipid metabolism, leading to the downregulation of a group of PPARα target genes involved in lipogenesis and decreased lipogenesis in the liver. Activating PPARα by fenofibrate, a PPARα agonist, restores lipid homeostasis in the liver and inhibits LIF-induced cachexia. These results provide valuable insights into cachexia, which may help develop strategies to treat cancer cachexia.


Asunto(s)
Caquexia , Neoplasias , Animales , Ratones , Caquexia/genética , Caquexia/metabolismo , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Lípidos , Lipogénesis/genética , Hígado/metabolismo , Ratones Transgénicos , Neoplasias/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo
10.
Fundam Clin Pharmacol ; 38(3): 596-605, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38192190

RESUMEN

BACKGROUND/OBJECTIVES: Clinical responses to naldemedine vary between individuals with advanced cancer. This is a prospective, single-center, observational study aimed to evaluate the influence of genetic polymorphisms and cachexia status on plasma naldemedine and clinical responses. METHODS: Forty-eight patients being treated with naldemedine for opioid-induced constipation under treatment of cancer pain were enrolled. Plasma naldemedine concentrations were determined on the fourth day or later after administration of naldemedine, and the associations with genotypes, cachexia status, and clinical responses were assessed. RESULTS: Cancer patients exhibited a large variation in the plasma naldemedine concentrations, and it was correlated with serum total protein level. Patients who were homozygous CYP3A5*3 had a higher plasma concentration of naldemedine than those with the *1 allele. ABCB1 genotypes tested in this study were not associated with plasma naldemedine. A negative correlation was observed between the plasma naldemedine concentration and 4ß-hydroxycholesterol level. The plasma naldemedine concentration was lower in patients with refractory cachexia than in those with precachexia and cachexia. While serum levels of interleukin-6 (IL-6) and acute-phase proteins were higher in patients with refractory cachexia, they were not associated with plasma naldemedine. A higher plasma concentration of naldemedine, CYP3A5*3/*3, and an earlier naldemedine administration after starting opioid analgesics were related to improvement of bowel movements. CONCLUSION: Plasma naldemedine increased under deficient activity of CYP3A5 in cancer patients. Cachectic patients with a higher serum IL-6 had a lower plasma naldemedine. Plasma naldemedine, related to CYP3A5 genotype, and the initiation timing of naldemedine were associated with improved bowel movements.


Asunto(s)
Analgésicos Opioides , Caquexia , Dolor en Cáncer , Citocromo P-450 CYP3A , Naltrexona , Polimorfismo Genético , Humanos , Masculino , Femenino , Caquexia/genética , Caquexia/tratamiento farmacológico , Caquexia/etiología , Persona de Mediana Edad , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/administración & dosificación , Naltrexona/análogos & derivados , Naltrexona/farmacocinética , Naltrexona/uso terapéutico , Naltrexona/efectos adversos , Estudios Prospectivos , Anciano , Citocromo P-450 CYP3A/genética , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/complicaciones , Genotipo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adulto , Estreñimiento Inducido por Opioides/genética , Estreñimiento Inducido por Opioides/tratamiento farmacológico , Defecación/efectos de los fármacos
11.
J Cachexia Sarcopenia Muscle ; 15(1): 149-158, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38123146

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is highly associated with cachexia and weight loss, which is driven by the tumour's effect on the body. Data are lacking on differences in these metrics based on PDAC anatomic location. We hypothesize that the primary tumour's anatomic region influences the prevalence and severity of unintentional weight loss. METHODS: Treatment naïve patients with PDAC who underwent pancreatectomy at a single institution between 2012 and 2020 were identified retrospectively. Patients with pancreatic head or distal tumours were matched by sex, age, N and T stage. Serologic and anthropometric variables were obtained at the time of diagnosis. Skeletal muscle index (SMI), muscle radiation attenuation (MRA) and adiposity were measured. The primary outcome was presence of significant weight loss [>5% body weight (BW) loss in past 6 months]. Signed rank tests, Cochran Mantel Haenszel tests and Kaplan-Meier survival analysis are presented. RNA-seq of tumours was performed to explore enriched pathways related to cachexia and weight loss. RESULTS: Pancreatic head tumours (n = 24) were associated with higher prevalence (70.8% vs. 41.7%, P = 0.081) and degree of weight loss (7.9% vs. 2.5%, P = 0.014) compared to distal tumours (n = 24). BMI (P = 0.642), SMI (P = 0.738) and MRA (P = 0.478) were similar between groups. Combining BW loss, SMI and MRA into a composite score, patients with pancreatic head cancers met more criteria associated with poor prognosis (P = 0.142). Serum albumin (3.9 vs. 4.4 g/dL, P = 0.002) was lower and bilirubin (4.5 vs. 0.4 mg/dL, P < 0.001) were higher with pancreatic head tumours. Survival differed by tumour location (P = 0.014) with numerically higher median overall survival with distal tumours (11.1 vs. 21.8 months; P = 0.066). Transcriptomic analysis revealed inactivation of appetite stimulation, weight regulation and nutrient digestion/metabolism pathways in pancreatic head tumours. CONCLUSIONS: Resectable pancreatic head PDAC is associated with higher prevalence of significant weight loss and more poor prognosis features. Pancreaticobiliary obstruction and hypoalbuminemia in patients with head tumours suggests compounding effects of nutrient malabsorption and systemic inflammation on molecular drivers of cachexia, possibly contributing to shorter survival. Therefore, PDAC-associated cachexia is a heterogenous syndrome, which may be influenced by the primary tumour location. Select patients with resectable pancreatic head tumours may benefit from nutritional rehabilitation to improve outcomes.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias de Cabeza y Cuello , Neoplasias Pancreáticas , Humanos , Caquexia/genética , Caquexia/complicaciones , Estudios Retrospectivos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Perfilación de la Expresión Génica , Neoplasias de Cabeza y Cuello/complicaciones
12.
Lipids Health Dis ; 22(1): 211, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38041133

RESUMEN

Cardiac cachexia is a deadly consequence of advanced heart failure that is characterised by the dysregulation of adipose tissue homeostasis. Once cachexia occurs with heart failure, it prevents the normal treatment of heart failure and increases the risk of death. Targeting adipose tissue is an important approach to treating cardiac cachexia, but the pathogenic mechanisms are still unknown, and there are no effective therapies available. Transcriptomics, metabolomics, and lipidomics were used to examine the underlying mechanisms of cardiac cachexia. Transcriptomics investigation of cardiac cachexia adipose tissue revealed that genes involved in fibrosis and monocyte/macrophage migration were increased and strongly interacted. The ECM-receptor interaction pathway was primarily enriched, as shown by KEGG enrichment analysis. In addition, gene set enrichment analysis revealed that monocyte chemotaxis/macrophage migration and fibrosis gene sets were upregulated in cardiac cachexia. Metabolomics enrichment analysis demonstrated that the sphingolipid signalling pathway is important for adipose tissue remodelling in cardiac cachexia. Lipidomics analysis showed that the adipose tissue of rats with cardiac cachexia had higher levels of sphingolipids, including Cer and S1P. Moreover, combined multiomics analysis suggested that the sphingolipid metabolic pathway was associated with inflammatory-fibrotic changes in adipose tissue. Finally, the key indicators were validated by experiments. In conclusion, this study described a mechanism by which the sphingolipid signalling pathway was involved in adipose tissue remodelling by inducing inflammation and fat fibrosis in cardiac cachexia.


Asunto(s)
Caquexia , Insuficiencia Cardíaca , Ratas , Animales , Caquexia/genética , Caquexia/complicaciones , Esfingolípidos/metabolismo , Multiómica , Tejido Adiposo/metabolismo , Fibrosis , Insuficiencia Cardíaca/patología , Obesidad/metabolismo
13.
Support Care Cancer ; 31(12): 708, 2023 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-37978991

RESUMEN

BACKGROUND: Malnutrition and cachexia are common syndromes in patients with gastric cancer (GC) and are associated with poor quality of life and poor disease prognosis. However, there is still a lack of molecular factors that can predict malnutrition or cachexia in cancer. Studies have shown that among the potential contributors to the development of cancer cachexia, the level of the inflammatory response to P-selectin is regulated by single nucleotide polymorphisms (SNPs) located in the promoter region of the SELP gene. The aim of this study was to evaluate the association between the single nucleotide polymorphism (SNP)-2028 A/G of the SELP gene and malnutrition in patients receiving chemotherapy for gastric cancer (GC). METHODS: The study group consisted of 220 GC patients treated with chemotherapy at Jinhua Municipal Central Hospital. DNA was extracted from peripheral leukocytes of whole blood samples using an animal DNA extraction kit. DNA was amplified using a 1.1 × T3 Super PCR mix, and loci corresponding to the peaks were genotyped using SNP1 software. RESULTS: Patients carrying the A allele had a reduced risk of developing malnutrition compared to patients with the GG genotype (P < 0.001; OR = 3.411; 95% CI = 1.785-6.516). In addition, multivariate analysis indicated that the AA genotype significantly (more than 16-fold) reduced the risk of developing malnutrition (P < 0.001; OR = 0.062; 95% CI = 0.015-0.255). CONCLUSION: SELP -2028A/G SNP may be a useful marker for assessing the risk of malnutrition in GC patients.


Asunto(s)
Desnutrición , Neoplasias Gástricas , Animales , Humanos , Caquexia/genética , Estudios de Casos y Controles , ADN , Predisposición Genética a la Enfermedad , Genotipo , Desnutrición/genética , Polimorfismo de Nucleótido Simple , Calidad de Vida , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética
14.
Front Immunol ; 14: 1297363, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38035067

RESUMEN

Cachexia, a debilitating condition that worsens patient outcomes, often accompanies gastric cancer, a malignancy that is prevalent worldwide. The extensive research explored the interconnected molecular and immune aspects of stomach cancer, with a particular emphasis on cachexia. By employing the GEO database, we identified genes that were expressed differently in gastric cancer patients suffering from cachexia. Following the analysis of Weighted Gene Co-expression Network (WGCNA), gene modules intricately linked to particular immune cells were revealed, indicating a significantly disrupted tumor microenvironment. A strong predictive model was developed, centered around key genes such as CAMK4, SLC37A2, and BCL11B. Surprisingly, this particular model not only showed better predictive abilities in comparison to conventional clinical factors but also exhibited a strong connection with increased infiltration of macrophages and T cells. These discoveries suggest the presence of an immune-suppressing and tumor-promoting atmosphere among individuals at a greater risk. Moreover, the utilization of Gene Set Enrichment Analysis (GSEA) established a connection between the genes linked to our risk score and vital immune-related pathways, thereby strengthening the pivotal involvement of immunity in the development of gastric cancer. To summarize, our discoveries provide a more profound comprehension of the molecular and immune mechanisms that support cachexia in gastric cancer, presenting a hopeful basis for upcoming advancements in treatment.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Pronóstico , Caquexia/genética , Bases de Datos Factuales , Redes Reguladoras de Genes , Microambiente Tumoral/genética , Proteínas Represoras , Proteínas Supresoras de Tumor
16.
Cell Rep ; 42(10): 113245, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37837622

RESUMEN

Many tumors recapitulate the developmental and differentiation program of their tissue of origin, a basis for tumor cell heterogeneity. Although stem-cell-like tumor cells are well studied, the roles of tumor cells undergoing differentiation remain to be elucidated. We employ Drosophila genetics to demonstrate that the differentiation program of intestinal stem cells is crucial for enabling intestinal tumors to invade and induce non-tumor-autonomous phenotypes. The differentiation program that generates absorptive cells is aberrantly recapitulated in the intestinal tumors generated by activation of the Yap1 ortholog Yorkie. Inhibiting it allows stem-cell-like tumor cells to grow but suppresses invasiveness and reshapes various phenotypes associated with cachexia-like wasting by altering the expression of tumor-derived factors. Our study provides insight into how a native differentiation program determines a tumor's capacity to induce advanced cancer phenotypes and suggests that manipulating the differentiation programs co-opted in tumors might alleviate complications of cancer, including cachexia.


Asunto(s)
Drosophila , Neoplasias Intestinales , Animales , Caquexia/genética , Diferenciación Celular/genética , Intestinos/patología , Neoplasias Intestinales/genética
17.
Genes (Basel) ; 14(9)2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37761958

RESUMEN

Previous studies have shown that inhibition of TNF family member FN14 (gene: TNFRSF12A) in colon tumors decreases inflammatory cytokine expression and mitigates cancer-induced cachexia. However, the molecular mechanisms underlying the regulation of FN14 expression remain unclear. Tumor microenvironments are often devoid of nutrients and oxygen, yet how the cachexic response relates to the tumor microenvironment and, importantly, nutrient stress is unknown. Here, we looked at the connections between metabolic stress and FN14 expression. We found that TNFRSF12A expression was transcriptionally induced during glutamine deprivation in cancer cell lines. We also show that the downstream glutaminolysis metabolite, alpha-ketoglutarate (aKG), is sufficient to rescue glutamine-deprivation-promoted TNFRSF12A induction. As aKG is a co-factor for histone de-methylase, we looked at histone methylation and found that histone H3K4me3 at the Tnfrsf12a promoter is increased under glutamine-deprived conditions and rescued via DM-aKG supplementation. Finally, expression of Tnfrsf12a and cachexia-induced weight loss can be inhibited in vivo by DM-aKG in a mouse cancer cachexia model. These findings highlight a connection between metabolic stress and cancer cachexia development.


Asunto(s)
Caquexia , Neoplasias del Colon , Receptor de TWEAK , Animales , Ratones , Caquexia/genética , Caquexia/prevención & control , Modelos Animales de Enfermedad , Glutamina/farmacología , Código de Histonas , Histona Metiltransferasas , Histonas/genética , Ácidos Cetoglutáricos/farmacología , Microambiente Tumoral , Humanos , Línea Celular Tumoral/metabolismo , Receptor de TWEAK/genética , Receptor de TWEAK/metabolismo
18.
Commun Biol ; 6(1): 977, 2023 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-37741882

RESUMEN

Cancer cachexia is characterized by weight loss and skeletal muscle wasting. Based on the up-regulation of catabolism and down-regulation of anabolism, here we showed genetic mutation-mediated metabolic reprogramming in the progression of cancer cachexia by screening for metabolites and investigating their direct effect on muscle atrophy. Treatment with 93 µM D-2-hydroxyglutarate (D2HG) resulted in reduced myotube width and increased expression of E3 ubiquitin ligases. Isocitrate Dehydrogenase 1 (IDH1) mutant patients had higher D2HG than non-mutant patients. In the in vivo murine cancer cachexia model, mutant IDH1 in CT26 cancer cells accelerated cachexia progression and worsened overall survival. Transcriptomics and metabolomics revealed a distinct D2HG-induced metabolic imbalance. Treatment with the IDH1 inhibitor ivosidenib delayed the progression of cancer cachexia in murine GL261 glioma model and CT26 colorectal carcinoma models. These data demonstrate the contribution of IDH1 mutation mediated D2HG accumulation to the progression of cancer cachexia and highlight the individualized treatment of IDH1 mutation associated cancer cachexia.


Asunto(s)
Caquexia , Glioma , Humanos , Animales , Ratones , Caquexia/genética , Caquexia/metabolismo , Atrofia Muscular/genética , Glioma/metabolismo , Fibras Musculares Esqueléticas/patología
19.
Proc Natl Acad Sci U S A ; 120(34): e2215095120, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37585460

RESUMEN

Cancer cachexia, and its associated complications, represent a large and currently untreatable roadblock to effective cancer management. Many potential therapies have been proposed and tested-including appetite stimulants, targeted cytokine blockers, and nutritional supplementation-yet highly effective therapies are lacking. Innovative approaches to treating cancer cachexia are needed. Members of the Kruppel-like factor (KLF) family play wide-ranging and important roles in the development, maintenance, and metabolism of skeletal muscle. Within the KLF family, we identified KLF10 upregulation in a multitude of wasting contexts-including in pancreatic, lung, and colon cancer mouse models as well as in human patients. We subsequently interrogated loss-of-function of KLF10 as a potential strategy to mitigate cancer associated muscle wasting. In vivo studies leveraging orthotopic implantation of pancreas cancer cells into wild-type and KLF10 KO mice revealed significant preservation of lean mass and robust suppression of pro-atrophy muscle-specific ubiquitin ligases Trim63 and Fbxo32, as well as other factors implicated in atrophy, calcium signaling, and autophagy. Bioinformatics analyses identified Transforming growth factor beta (TGF-ß), a known inducer of KLF10 and cachexia promoting factor, as a key upstream regulator of KLF10. We provide direct in vivo evidence that KLF10 KO mice are resistant to the atrophic effects of TGF-ß. ChIP-based binding studies demonstrated direct binding to Trim63, a known wasting-associated atrogene. Taken together, we report a critical role for the TGF-ß/KLF10 axis in the etiology of pancreatic cancer-associated muscle wasting and highlight the utility of targeting KLF10 as a strategy to prevent muscle wasting and limit cancer-associated cachexia.


Asunto(s)
Neoplasias Pancreáticas , Factor de Crecimiento Transformador beta , Humanos , Ratones , Animales , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Caquexia/genética , Atrofia Muscular/genética , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Músculo Esquelético/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo
20.
Nat Commun ; 14(1): 5179, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37620316

RESUMEN

Cancer-associated cachexia is a multi-organ weight loss syndrome, especially with a wasting disorder of adipose tissue and skeletal muscle. Small extracellular vesicles (sEVs) serve as emerging messengers to connect primary tumour and metabolic organs to exert systemic regulation. However, whether and how tumour-derived sEVs regulate white adipose tissue (WAT) browning and fat loss is poorly defined. Here, we report breast cancer cell-secreted exosomal miR-204-5p induces hypoxia-inducible factor 1A (HIF1A) in WAT by targeting von Hippel-Lindau (VHL) gene. Elevated HIF1A protein induces the leptin signalling pathway and thereby enhances lipolysis in WAT. Additionally, exogenous VHL expression blocks the effect of exosomal miR-204-5p on WAT browning. Reduced plasma phosphatidyl ethanolamine level is detected in mice lack of cancer-derived miR-204-5p secretion in vivo. Collectively, our study reveals circulating miR-204-5p induces hypoxia-mediated leptin signalling pathway to promote lipolysis and WAT browning, shedding light on both preventive screenings and early intervention for cancer-associated cachexia.


Asunto(s)
Tejido Adiposo Blanco , Leptina , MicroARNs , Neoplasias , Animales , Ratones , Tejido Adiposo Blanco/metabolismo , Caquexia/genética , Hipoxia , MicroARNs/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA