Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nature ; 579(7797): 111-117, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32103177

RESUMO

The avascular nature of cartilage makes it a unique tissue1-4, but whether and how the absence of nutrient supply regulates chondrogenesis remain unknown. Here we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate forkhead box O (FOXO) transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing oxidation of fatty acids, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FOXO transcription factors during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.


Assuntos
Osso e Ossos/citologia , Microambiente Celular , Condrogênese , Metabolismo dos Lipídeos , Fatores de Transcrição SOX9/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Osso e Ossos/irrigação sanguínea , Condrócitos/citologia , Condrócitos/metabolismo , Ácidos Graxos/metabolismo , Feminino , Privação de Alimentos , Fatores de Transcrição Forkhead/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese , Oxirredução , Fatores de Transcrição SOX9/genética , Transdução de Sinais , Cicatrização
2.
Acta Neuropathol Commun ; 11(1): 11, 2023 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-36641470

RESUMO

Automatic segmentation of rodent brain tumor on magnetic resonance imaging (MRI) may facilitate biomedical research. The current study aims to prove the feasibility for automatic segmentation by artificial intelligence (AI), and practicability of AI-assisted segmentation. MRI images, including T2WI, T1WI and CE-T1WI, of brain tumor from 57 WAG/Rij rats in KU Leuven and 46 mice from the cancer imaging archive (TCIA) were collected. A 3D U-Net architecture was adopted for segmentation of tumor bearing brain and brain tumor. After training, these models were tested with both datasets after Gaussian noise addition. Reduction of inter-observer disparity by AI-assisted segmentation was also evaluated. The AI model segmented tumor-bearing brain well for both Leuven and TCIA datasets, with Dice similarity coefficients (DSCs) of 0.87 and 0.85 respectively. After noise addition, the performance remained unchanged when the signal-noise ratio (SNR) was higher than two or eight, respectively. For the segmentation of tumor lesions, AI-based model yielded DSCs of 0.70 and 0.61 for Leuven and TCIA datasets respectively. Similarly, the performance is uncompromised when the SNR was over two and eight respectively. AI-assisted segmentation could significantly reduce the inter-observer disparities and segmentation time in both rats and mice. Both AI models for segmenting brain or tumor lesions could improve inter-observer agreement and therefore contributed to the standardization of the following biomedical studies.


Assuntos
Inteligência Artificial , Neoplasias Encefálicas , Ratos , Camundongos , Animais , Processamento de Imagem Assistida por Computador/métodos , Roedores , Imageamento por Ressonância Magnética/métodos , Neoplasias Encefálicas/diagnóstico por imagem
3.
Cancers (Basel) ; 14(7)2022 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-35406378

RESUMO

The epidemiology and associated potential heterogeneity of synchronous lung metastasis (sLM) have not been reported at a population-based level. Cancer patients with valid information about sLM status in the Surveillance, Epidemiology, and End Results database were enrolled. The prevalence of sLM, with a 95% confidential interval, and median survival of sLM, with interquartile range, were calculated and compared by Chi-square analyses and log-rank tests by primary cancer type and clinicopathological factors. Furthermore, the risk factors of sLM development were identified by multivariate logistic regression. Among 1,672,265 enrolled cases, 3.3% cases were identified with sLM, with a median survival of 7 months. Heterogeneity in prevalence and prognosis in sLM was observed among different primary cancers, with the highest prevalence in main bronchus cancer and best survival in testis cancer. Higher prevalence and poorer prognosis were observed in the older population, male population, African American, patients with lower socioeconomic status, and cases with advanced T stage, N stage, or more malignant pathological characteristics. Race, age, T stage, N stage, metastasis to other sites, insurance status and marital status were associated with sLM development (p < 0.001). The current study highlights the heterogeneity of the prevalence and prognosis in patients with sLM.

4.
Nat Commun ; 13(1): 2760, 2022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35589749

RESUMO

Autophagy has vasculoprotective roles, but whether and how it regulates lymphatic endothelial cells (LEC) homeostasis and lymphangiogenesis is unknown. Here, we show that genetic deficiency of autophagy in LEC impairs responses to VEGF-C and injury-driven corneal lymphangiogenesis. Autophagy loss in LEC compromises the expression of main effectors of LEC identity, like VEGFR3, affects mitochondrial dynamics and causes an accumulation of lipid droplets (LDs) in vitro and in vivo. When lipophagy is impaired, mitochondrial ATP production, fatty acid oxidation, acetyl-CoA/CoA ratio and expression of lymphangiogenic PROX1 target genes are dwindled. Enforcing mitochondria fusion by silencing dynamin-related-protein 1 (DRP1) in autophagy-deficient LEC fails to restore LDs turnover and lymphatic gene expression, whereas supplementing the fatty acid precursor acetate rescues VEGFR3 levels and signaling, and lymphangiogenesis in LEC-Atg5-/- mice. Our findings reveal that lipophagy in LEC by supporting FAO, preserves a mitochondrial-PROX1 gene expression circuit that safeguards LEC responsiveness to lymphangiogenic mediators and lymphangiogenesis.


Assuntos
Linfangiogênese , Vasos Linfáticos , Animais , Autofagia/genética , Células Endoteliais/metabolismo , Ácidos Graxos/metabolismo , Gotículas Lipídicas/metabolismo , Linfangiogênese/genética , Vasos Linfáticos/metabolismo , Camundongos , Mitocôndrias , Fatores de Transcrição/metabolismo
5.
Nutrients ; 13(10)2021 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-34684461

RESUMO

(1) Background: Obesity and type 2 diabetes have been suspected to impact both intrinsic metabolism and function of circulating immune cells. (2) Methods: To further investigate this immunometabolic modulation, we profiled the phospholipidome of the peripheral blood mononuclear cells (PBMCs) in lean, normoglycemic obese (OBNG) and obese with dysglycemia (OBDysG) individuals. (3) Results: The global PBMCs phospholipidome is significantly downmodulated in OBDysG unlike OBNG patients when compared to lean ones. Multiple linear regression analyses show a strong negative relationship between the global PBMCs phospholipidome and parameters assessing insulin resistance. Even though all classes of phospholipid are affected, the relative abundance of each class is maintained with the exception of Lyso-PC/PC and Lyso-PE/PE ratios that are downmodulated in PBMCs of OBDysG compared to OBNG individuals. Interestingly, the percentage of saturated PC is positively associated with glycated hemoglobin (HbA1c). Moreover, a few lipid species are significantly downmodulated in PBMCs of OBDysG compared to OBNG individuals, making possible to distinguish the two phenotypes. (4) Conclusions: This lipidomic study highlights for the first-time modulations of the PBMCs phospholipidome in obese patients with prediabetes and type 2 diabetes. Such phospholipidome remodeling could disrupt the cell membranes and the lipid mediator's levels, driving an immune cell dysfunction.


Assuntos
Glicemia , Resistência à Insulina , Leucócitos Mononucleares/metabolismo , Lipidômica , Obesidade/metabolismo , Fosfolipídeos/metabolismo , Adulto , Biomarcadores , Pesos e Medidas Corporais , Biologia Computacional , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Lipidômica/métodos , Masculino , Espectrometria de Massas , Lipídeos de Membrana , Pessoa de Meia-Idade , Obesidade/sangue , Obesidade/etiologia , Adulto Jovem
6.
Cancer Metab ; 8: 11, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32577235

RESUMO

BACKGROUND: Metabolic reprograming, non-mutational epigenetic changes, increased cell plasticity, and multidrug tolerance are early hallmarks of therapy resistance in cancer. In this temporary, therapy-tolerant state, cancer cells are highly sensitive to ferroptosis, a form of regulated cell death that is caused by oxidative stress through excess levels of iron-dependent peroxidation of polyunsaturated fatty acids (PUFA). However, mechanisms underpinning therapy-induced ferroptosis hypersensitivity remain to be elucidated. METHODS: We used quantitative single-cell imaging of fluorescent metabolic probes, transcriptomics, proteomics, and lipidomics to perform a longitudinal analysis of the adaptive response to androgen receptor-targeted therapies (androgen deprivation and enzalutamide) in prostate cancer (PCa). RESULTS: We discovered that cessation of cell proliferation and a robust reduction in bioenergetic processes were associated with multidrug tolerance and a strong accumulation of lipids. The gain in lipid biomass was fueled by enhanced lipid uptake through cargo non-selective (macropinocytosis, tunneling nanotubes) and cargo-selective mechanisms (lipid transporters), whereas de novo lipid synthesis was strongly reduced. Enzalutamide induced extensive lipid remodeling of all major phospholipid classes at the expense of storage lipids, leading to increased desaturation and acyl chain length of membrane lipids. The rise in membrane PUFA levels enhanced membrane fluidity and lipid peroxidation, causing hypersensitivity to glutathione peroxidase (GPX4) inhibition and ferroptosis. Combination treatments against AR and fatty acid desaturation, lipase activities, or growth medium supplementation with antioxidants or PUFAs altered GPX4 dependence. CONCLUSIONS: Our work provides mechanistic insight into processes of lipid metabolism that underpin the acquisition of therapy-induced GPX4 dependence and ferroptosis hypersensitivity to standard of care therapies in PCa. It demonstrates novel strategies to suppress the therapy-tolerant state that may have potential to delay and combat resistance to androgen receptor-targeted therapies, a currently unmet clinical challenge of advanced PCa. Since enhanced GPX4 dependence is an adaptive phenotype shared by several types of cancer in response to different therapies, our work might have universal implications for our understanding of metabolic events that underpin resistance to cancer therapies.

7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(7): 1017-1030, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30953761

RESUMO

NLRP3 inflammasome plays a key role in Western diet-induced systemic inflammation and was recently shown to mediate long-lasting trained immunity in myeloid cells. Saturated fatty acids (SFAs) are sterile triggers able to induce the assembly of the NLRP3 inflammasome in macrophages, leading to IL-1ß secretion while unsaturated ones (UFAs) prevent SFAs-mediated NLRP3 activation. Unlike previous studies using LPS-primed bone marrow derived macrophages, we do not see any ROS or IRE-1α involvement in SFAs-mediated NLRP3 activation in human monocytes-derived macrophages. Rather we show that SFAs need to enter the cells and to be activated into acyl-CoA to lead to NLRP3 activation in human macrophages. However, their ß-oxidation is dispensable. Instead, they are channeled towards phospholipids but redirected towards lipid droplets containing triacylglycerol in the presence of UFAs. Lipidomic analyses and Laurdan fluorescence experiments demonstrate that SFAs induce a dramatic saturation of phosphatidylcholine (PC) correlated with a loss of membrane fluidity, both events inhibited by UFAs. The silencing of CCTα, the key enzyme in PC synthesis, prevents SFA-mediated NLRP3 activation, demonstrating the essential role of the de novo PC synthesis. This SFA-induced membrane remodeling promotes a disruption of the plasma membrane Na, K-ATPase, instigating a K+ efflux essential and sufficient for NLRP3 activation. This work opens novel therapeutic avenues to interfere with Western diet-associated diseases such as those targeting the glycerolipid pathway.


Assuntos
Ácidos Graxos/metabolismo , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Transporte Biológico , Células Cultivadas , Humanos , Inflamassomos/metabolismo , Fosfolipídeos/metabolismo
8.
Oncotarget ; 8(24): 38264-38275, 2017 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-28415728

RESUMO

Prostate cancer is the most common male cancer and androgen receptor (AR) is the major driver of the disease. Here we show that Enoyl-CoA delta isomerase 2 (ECI2) is a novel AR-target that promotes prostate cancer cell survival. Increased ECI2 expression predicts mortality in prostate cancer patients (p = 0.0086). ECI2 encodes for an enzyme involved in lipid metabolism, and we use multiple metabolite profiling platforms and RNA-seq to show that inhibition of ECI2 expression leads to decreased glucose utilization, accumulation of fatty acids and down-regulation of cell cycle related genes. In normal cells, decrease in fatty acid degradation is compensated by increased consumption of glucose, and here we demonstrate that prostate cancer cells are not able to respond to decreased fatty acid degradation. Instead, prostate cancer cells activate incomplete autophagy, which is followed by activation of the cell death response. Finally, we identified a clinically approved compound, perhexiline, which inhibits fatty acid degradation, and replicates the major findings for ECI2 knockdown. This work shows that prostate cancer cells require lipid degradation for survival and identifies a small molecule inhibitor with therapeutic potential.


Assuntos
Dodecenoil-CoA Isomerase/metabolismo , Regulação Neoplásica da Expressão Gênica , Metabolismo dos Lipídeos/fisiologia , Neoplasias da Próstata/metabolismo , Biomarcadores Tumorais/análise , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Estimativa de Kaplan-Meier , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Perexilina/farmacologia , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA