Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Life Sci ; 333: 122172, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37832632

RESUMO

AIMS: Cachexia, a metabolic syndrome, affects 21 % of patients suffering from ischemic encephalopathy. However, the specific mechanism and prevention measures are still unclear. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been proven to reduce inflammatory cytokine levels during ischemic events, but whether they have a protective effect against cachexia after hypoxic-ischemic brain damage (HIBD) remains unclear. MAIN METHODS: C57BL/6J wild-type and mfat-1 transgenic male mice were treated with and without HIBD. One day after HIBD, the epididymal white fat, gastrocnemius muscle and hypothalamus were weighed and analyzed the phenotypic changes. RNA sequencing was applied to gastrocnemius muscle to identify differential genes and pathways in HIBD groups. The effect of HPA axis on cachexia post-HIBD was examined via adrenalectomy, dexamethasone (0.1 mg/kg), and corticosterone injection (100 mg/kg). KEY FINDINGS: The results showed that the incidence of cachexia in mfat-1 mice, which produce high proportion of n-3 PUFAs, was significantly lower than that in wild-type mice post-HIBD. Cachexia-related factors, such as inflammation, muscle atrophy and lipid metabolism were significantly improved in mfat-1 HIBD. RNA sequencing revealed that catabolic and proteasome pathways were significantly downregulated. In hypothalamus, inflammatory cytokines, lipid peroxidation levels were reduced. Corticosterone, glucocorticoid receptor, and dexamethasone suppression test all showed that mfat-1 improved the dysfunction of the HPA axis post-HIBD. The present study elucidated for the first time that mfat-1 reduced HIBD-induced hyperactivation of the HPA axis in mice by reducing inflammation and oxidative stress and contributed to the reduction of metabolic imbalance in peripheral tissues. SIGNIFICANCE: Our study provides mechanistic information for the development of intervention strategies to prevent cachexia.


Assuntos
Sistema Hipotálamo-Hipofisário , Hipóxia-Isquemia Encefálica , Humanos , Camundongos , Animais , Masculino , Sistema Hipotálamo-Hipofisário/metabolismo , Caquexia/etiologia , Caquexia/prevenção & controle , Caquexia/metabolismo , Corticosterona/metabolismo , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Camundongos Transgênicos , Hipotálamo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Inflamação/metabolismo , Dexametasona/metabolismo , Animais Recém-Nascidos , Encéfalo/metabolismo
2.
Biomed Pharmacother ; 167: 115507, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37722192

RESUMO

Phenylethanoid glycosides derived from Cistanche deserticola (PhGs) are plant-derived natural medicinal compounds that occur in many medicinal plants. This study aims to investigate whether PhGs treatment improves the stroke and its potential mechanisms. Adult male C57BL/6 J mice were administrated PhGs once daily for 7 days after MCAO surgery. The neurological score, and catwalk were evaluated on Day 1, 3 and 7 after ischemic stroke. Furthermore, triphenyl-2,3,5-tetrazoliumchloride (TTC) and hematoxylin-eosin (H&E) staining were used for evaluating the infarct volume and neuronal restoration. The effects of PhGs on NSCs proliferation were investigated in vitro and in vivo. Western blot was used to detect the proteins of Wnt/ß-catenin signaling pathway. This study found that PhGs effectively improved the neurological functions in ischemic stroke mice. TTC and H&E staining demonstrated that PhGs not only reduced infarct volume, but also improved neuronal restoration. The immunohistochemistry and 5-Ethynyl-2-deoxyuridine (EdU) incorporation assays revealed that PhGs promoted the proliferation of neural stem cells (NSCs) in subventricular zone (SVZ). In addition, transcriptome analysis of NSCs showed that the Wnt/ß-catenin signaling pathway was involved in the PhGs induced NSCs proliferation. Importantly, the related proteins in the Wnt/ß-catenin signaling pathway were changed after PhGs treatment, including ß-catenin, Wnt3a, GSK-3ß, c-Myc. PhGs treatment improved the stroke through enhancing endogenous NSCs proliferation via activating Wnt/ß-catenin signaling pathway. Due to its effect on the proliferation of NSCs, PhGs are a potential adjuvant therapeutic drug for post-stroke treatment.

3.
Nat Commun ; 11(1): 5465, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33122660

RESUMO

Eicosapentaenoic acid (EPA), an omega-3 fatty acid, has been widely used to prevent cardiovascular disease (CVD) and treat brain diseases alone or in combination with docosahexaenoic acid (DHA). However, the impact of EPA and DHA supplementation on normal cognitive function and the molecular targets of EPA and DHA are still unknown. We show that acute administration of EPA impairs learning and memory and hippocampal LTP in adult and prepubescent mice. Similar deficits are duplicated by endogenously elevating EPA in the hippocampus in the transgenic fat-1 mouse. Furthermore, the damaging effects of EPA are mediated through enhancing GABAergic transmission via the 5-HT6R. Interestingly, DHA can prevent EPA-induced impairments at a ratio of EPA to DHA similar to that in marine fish oil via the 5-HT2CR. We conclude that EPA exhibits an unexpected detrimental impact on cognitive functions, suggesting that caution must be exercised in omega-3 fatty acid supplementation and the combination of EPA and DHA at a natural ratio is critical for learning and memory and synaptic plasticity.


Assuntos
Cognição/efeitos dos fármacos , Ácido Eicosapentaenoico/efeitos adversos , Neurônios GABAérgicos/efeitos dos fármacos , Receptor 5-HT2C de Serotonina/efeitos dos fármacos , Animais , Suplementos Nutricionais/efeitos adversos , Ácidos Docosa-Hexaenoicos/farmacologia , Combinação de Medicamentos , Ácido Eicosapentaenoico/farmacologia , Ácidos Graxos Ômega-3/efeitos adversos , Óleos de Peixe/efeitos adversos , Óleos de Peixe/farmacologia , Humanos , Aprendizagem/efeitos dos fármacos , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Camundongos
4.
Bioengineered ; 10(1): 397-408, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31526157

RESUMO

Nowadays, there is an increasing concern toward substituting the scarce wood fibers with alternative lignocellulosic fibers that originate from crop residue to reinforce biocomposites. In this paper, the potential application of coffee hull (CH) of the reinforced polyethylene (PE) matrix composites was studied for the first time. Experiments of composite that enhanced with CH on mechanical properties, hydroscopicity, thermogravimetric analysis, fiber treatment, and microstructures were tested in this study. The PE matrix was reinforced with varying volume fractions of CH and was studied. The results show that incorporation of coffee hull markedly improved the mechanical properties of the reinforced high-density polyethylene (HDPE) matrix composites. Micrographs show a strong interfacial adhesion between the CH fiber particles. This property may be the main reason for the stability between composites. At the same time this work investigated the effect of different treatments on the mechanical properties and water absorption behavior of composites. The fiber surface treatments were done using active chemicals such as calcium hydroxide (Ca(OH)2), silane coupling agent (SCA), maleic anhydride grafted polypropylene (MA-g-PP), stearic acid (SA), ethylene bis stearamide (EBS) and the combination (MA-g-PP, SA, EBS). The results show that (Ca(OH)2)treatment is the best way to improve its properties. Probably because attributed to removal of surface active functional groups (-OH) from the CH fiber and induction of hydrophobicity that in turn improved the compatibility with the polymer matrix. As a result, the use of coffee hull in composites could have great significance for the industry.


Assuntos
Café/química , Manufaturas , Plásticos/síntese química , Polietileno/química , Hidróxido de Cálcio/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Anidridos Maleicos/química , Teste de Materiais , Plásticos/química , Silanos/química , Ácidos Esteáricos/química , Temperatura , Resistência à Tração , Resíduos , Molhabilidade
5.
Sci Rep ; 7: 46146, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28393852

RESUMO

Epidemiologic studies showed the correlation between the deficiency of omega-3 polyunsaturated fatty acids (n-3 PUFAs) and the progression of chronic kidney diseases (CKD), however, the role and mechanisms for n-3 PUFAs in protecting against kidney fibrosis remain obscure. In this study, NRK-49F cells, a rat kidney interstitial fibroblast cell line, were stimulated with TGFß1. A Caenorhabditis elegans fat-1 transgenic mouse model in which n-3 PUFAs are endogenously produced from n-6 PUFAs owing to the expression of n-3 fatty acid desaturase were deployed. Docosahexaenoic acid (DHA), one member of n-3 PUFAs family, could suppress TGFß1-induced fibroblast activation at a dose and time dependent manner. Additionally, DHA could largely inhibit TGFß1-stimulated Akt but not S6 or Smad3 phosphorylation at a time dependent manner. To decipher the role for n-3 PUFAs in protecting against kidney fibrosis, fat-1 transgenic mice were operated with unilateral ureter obstruction (UUO). Compared to the wild types, fat-1 transgenics developed much less kidney fibrosis and inflammatory cell accumulation accompanied by less p-Akt (Ser473), p-Akt (Thr308), p-S6 and p-Smad3 in kidney tissues at day 7 after UUO. Thus, n-3 PUFAs can attenuate fibroblast activation and kidney fibrosis, which may be associated with the inhibition of mTORC2 signaling.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Fibroblastos/patologia , Rim/patologia , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Linhagem Celular , Matriz Extracelular/metabolismo , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Ômega-3/uso terapêutico , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Inflamação/patologia , Nefropatias/patologia , Nefropatias/terapia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Fator de Crescimento Transformador beta1/farmacologia , Transgenes , Obstrução Ureteral/tratamento farmacológico , Obstrução Ureteral/patologia
6.
J Clin Invest ; 127(5): 1757-1771, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28375156

RESUMO

Despite the benefit of insulin, blockade of autoimmune attack and regeneration of pancreatic islets are ultimate goals for the complete cure of type 1 diabetes (T1D). Long-term consumption of ω-3 polyunsaturated fatty acids (PUFAs) is known to suppress inflammatory processes, making these fatty acids candidates for the prevention and amelioration of autoimmune diseases. Here, we explored the preventative and therapeutic effects of ω-3 PUFAs on T1D. In NOD mice, dietary intervention with ω-3 PUFAs sharply reduced the incidence of T1D, modulated the differentiation of Th cells and Tregs, and decreased the levels of IFN-γ, IL-17, IL-6, and TNF-α. ω-3 PUFAs exerted similar effects on the differentiation of CD4+ T cells isolated from human peripheral blood mononuclear cells. The regulation of CD4+ T cell differentiation was mediated at least in part through ω-3 PUFA eicosanoid derivatives and by mTOR complex 1 (mTORC1) inhibition. Importantly, therapeutic intervention in NOD mice through nutritional supplementation or lentivirus-mediated expression of an ω-3 fatty acid desaturase, mfat-1, normalized blood glucose and insulin levels for at least 182 days, blocked the development of autoimmunity, prevented lymphocyte infiltration into regenerated islets, and sharply elevated the expression of the ß cell markers pancreatic and duodenal homeobox 1 (Pdx1) and paired box 4 (Pax4). The findings suggest that ω-3 PUFAs could potentially serve as a therapeutic modality for T1D.


Assuntos
Autoimunidade/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Ácidos Graxos Ômega-3/farmacologia , Animais , Autoimunidade/genética , Glicemia/genética , Glicemia/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Caderinas/genética , Caderinas/imunologia , Citocinas/genética , Citocinas/imunologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos NOD , Complexos Multiproteicos/genética , Complexos Multiproteicos/imunologia , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/imunologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia , Transativadores/genética , Transativadores/imunologia
7.
Oncotarget ; 7(47): 76944-76954, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27769066

RESUMO

Although epidemiological and preclinical studies have shown the preventative effect of n-3 polyunsaturated fatty acids (PUFAs) on colorectal cancer (CRC), the underlying molecular mechanisms are not clear. In this study, we revealed that elevation of n-3/n-6 PUFAs ratio suppress the mechanistic target of rapamycin complex 1 (mTORC1) and prevent colorectal tumorigenesis. The transgenic expression of fat-1, a desaturase that catalyzes the conversion of n-6 to n-3 PUFAs and produces n-3 PUFAs endogenously, repressed colorectal tumor cell growth and remarkably reduced tumor burden, and alleviated anemia as well as hyperlipidemia in APCMin/+ (adenomatous polyposis coli) mice, a classic CRC model that best simulates most clinical cases. In contrast to arachidonic acid (AA, C20:4 n-6), either Docosahexaenoic acid (DHA, C22:6 n-3), eicosapentaenoic acid (EPA, C20:5 n-3), or a combination of DHA and AA, efficiently inhibited the proliferation of CRC cell lines and promoted apoptosis in these cells. The ectopic expression of fat-1 had similar effects in colon epithelial cells with APC depletion. Mechanistically, elevation of n-3/n-6 ratio suppressed mTORC1 activity in tumors of APCMin/+ mice, CRC cell lines with APC mutation, and in normal colon epithelial cells with APC depletion. In addition, elevation of n-3/n-6 ratio repressed mTORC1 activity and inhibited adipogenic differentiation in preadipocytes with APC knockdown, as well as alleviated hyperlipidemia in APCMin/+ mice. Taken together, our findings have provided novel insights into the potential mechanism by which increase in n-3/n-6 PUFAs ratio represses CRC development, and also a new rationale for utilizing n-3 PUFAs in CRC prevention and treatment.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias Colorretais/prevenção & controle , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Ômega-3/administração & dosagem , Serina-Treonina Quinases TOR/genética , Células 3T3-L1 , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/genética , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-6/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Neoplasias Experimentais/genética , Neoplasias Experimentais/prevenção & controle
8.
Mol Endocrinol ; 28(8): 1316-28, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24978197

RESUMO

Despite the well-documented health benefits of ω-3 polyunsaturated fatty acids (PUFAs), their use in clinical management of hyperglycemia and obesity has shown little success. To better define the mechanisms of ω-3 PUFAs in regulating energy balance and insulin sensitivity, we deployed a transgenic mouse model capable of endogenously producing ω-3 PUFAs while reducing ω-6 PUFAs owing to the expression of a Caenorhabditis elegans fat-1 gene encoding an ω-3 fatty acid desaturase. When challenged with high-fat diets, fat-1 mice strongly resisted obesity, diabetes, hypercholesterolemia, and hepatic steatosis. Endogenous elevation of ω-3 PUFAs and reduction of ω-6 PUFAs did not alter the amount of food intake but led to increased energy expenditure in the fat-1 mice. The requirements for the levels of ω-3 PUFAs as well as the ω-6/ω-3 ratios in controlling blood glucose and obesity are much more stringent than those in lipid metabolism. These metabolic phenotypes were accompanied by attenuation of the inflammatory state because tissue levels of prostaglandin E2, leukotriene B4, monocyte chemoattractant protein-1, and TNF-α were significantly decreased. TNF-α-induced nuclear factor-κB signaling was almost completely abolished. Consistent with the reduction in chronic inflammation and a significant increase in peroxisome proliferator-activated receptor-γ activity in the fat-1 liver tissue, hepatic insulin signaling was sharply elevated. The activities of prolipogenic regulators, such as liver X receptor, stearoyl-CoA desaturase-1, and sterol regulatory element binding protein-1 were sharply decreased, whereas the activity of peroxisome proliferator-activated receptor-α, a nuclear receptor that facilitates lipid ß-oxidation, was markedly increased. Thus, endogenous conversion of ω-6 to ω-3 PUFAs via fat-1 strongly protects against obesity, diabetes, inflammation, and dyslipidemia and may represent a novel therapeutic modality to treat these prevalent disorders.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Dislipidemias/metabolismo , Ácidos Graxos Ômega-3/biossíntese , Obesidade/metabolismo , Adiposidade , Animais , Caderinas/genética , Caderinas/metabolismo , Diabetes Mellitus Experimental/etiologia , Dieta Hiperlipídica/efeitos adversos , Resistência à Doença , Dislipidemias/etiologia , Expressão Gênica , Inflamação , Lipogênese , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/etiologia
9.
Cancer Prev Res (Phila) ; 7(8): 824-34, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24866178

RESUMO

Although preclinical and epidemiologic studies have shown the importance of n-3 polyunsaturated fatty acids (PUFA) in the prevention of hormone-responsive cancers such as breast cancer, evidence of the association between n-3 PUFAs and endometrial cancer risk is limited and no previous study has examined the effect of n-3 PUFAs on endometrial cancer in cellular and animal models. In this study, we demonstrated that docosahexenoic acid (DHA) dose- and time-dependently inhibited endometrial cancer cell proliferation, colony formation, and migration and promoted apoptosis. Dietary n-3 PUFAs efficiently prevented endometrial cancer cell growth in xenograft models. Moreover, ectopic expression of fat-1, a desaturase, catalyzed the conversion of n-6 to n-3 PUFAs and produced n-3 PUFAs endogenously, also suppressed endometrial tumor cell growth and migration, and potentiated apoptosis in endometrial cancer cell lines. Interestingly, implanted endometrial cancer cells were unable to grow in fat-1 transgenic SCID mice. Further study revealed that mTOR signaling, which plays an essential role in cell proliferation and endometrial tumorigenesis, is a target of n-3 PUFAs. Exogenous or endogenous n-3 PUFAs efficiently suppressed both mTOR complex 1 (mTORC1) and mTORC2 in vitro and in vivo. Moreover, both dietary n-3 PUFAs and transgenic expression of fat-1 in mice effectively repressed mTORC1/2 signaling and endometrial growth elicited by unopposed estrogen. Taken together, our findings provide comprehensive preclinical evidences that n-3 PUFAs efficiently prevent endometrial cancer and establish mTORC1/2 as a target of n-3 PUFAs.


Assuntos
Dieta , Neoplasias do Endométrio/tratamento farmacológico , Ácidos Graxos Ômega-3/administração & dosagem , Animais , Apoptose , Catálise , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Complexos Multiproteicos/metabolismo , Transplante de Neoplasias , Serina-Treonina Quinases TOR/metabolismo , Tamoxifeno/química , Cicatrização
10.
Ann Rheum Dis ; 73(9): 1719-27, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23852692

RESUMO

BACKGROUND: An exogenous supplement of n-3 polyunsaturated fatty acids (PUFAs) has been reported to prevent osteoarthritis (OA) through undefined mechanisms. OBJECTIVE: This study investigated the effect of alterations in the composition of endogenous PUFAs on OA, and associations of PUFAs with mammalian target of rapamycin complex 1 (mTORC1) signalling, a critical autophagy pathway in fat-1 transgenic (TG) mice. METHODS: fat-1 TG and wild-type mice were used to create an OA model by resecting the medial meniscus. The composition of the endogenous PUFAs in mouse tissues was analysed by gas chromatography, and the incidence of OA was evaluated by micro-computed tomography (micro-CT), scanning electron microscopy and histological methods. Additionally, primary chondrocytes were isolated and cultured. The effect of exogenous and endogenous PUFAs on mTORC1 activity and autophagy in chondrocytes was assessed. RESULTS: The composition of endogenous PUFAs of TG mice was optimised both by increased n-3 PUFAs and decreased n-6 PUFAs, which significantly alleviated the articular cartilage destruction and osteophytosis in the OA model (p<0.01), decreased protein expression of matrix metalloproteinase-13 (MMP-13) and ADAMTS-5 (a disintegrin and metalloproteinase with thrombospondin motifs) in the articular cartilage (p<0.01) and reduced chondrocyte number and loss of cartilage extracellular matrix. Both exogenous and endogenous n-3 PUFAs downregulated mTORC1 activity and promoted autophagy in articular chondrocytes. Conversely, mTORC1 pathway activation suppressed autophagy in articular chondrocytes. CONCLUSIONS: Enhancement of the synthesis of endogenous n-3 PUFAs from n-6 PUFAs can delay the incidence of OA, probably through inhibition of mTORC1, promotion of autophagy and cell survival in cartilage chondrocytes. Future investigation into the role of the endogenous n-6/n-3 PUFAs composition in OA prevention and treatment is warranted.


Assuntos
Artrite Experimental/prevenção & controle , Ácidos Graxos Ômega-3/biossíntese , Complexos Multiproteicos/fisiologia , Osteoartrite/prevenção & controle , Serina-Treonina Quinases TOR/fisiologia , Proteínas ADAM/metabolismo , Proteína ADAMTS5 , Animais , Artrite Experimental/etiologia , Artrite Experimental/patologia , Autofagia/fisiologia , Caderinas/genética , Cartilagem Articular/metabolismo , Cartilagem Articular/ultraestrutura , Condrócitos/patologia , Progressão da Doença , Ácidos Graxos Ômega-3/fisiologia , Ácidos Graxos Ômega-6/biossíntese , Feminino , Metaloproteinase 13 da Matriz/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Osteoartrite/etiologia , Osteoartrite/patologia , Proteoglicanas/metabolismo , Transdução de Sinais/fisiologia
11.
Drug Des Devel Ther ; 7: 545-52, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23843691

RESUMO

AIM: To investigate the effect of endogenous n-3 polyunsaturated fatty acids (PUFAs) on bone marrow adipogenesis under osteoporosis conditions. METHODS: A mouse osteoporosis model overexpressing the FAT1 gene from Caenorhabditis elegans and converting n-6 PUFAs to n-3 PUFAs endogenously was used. RESULTS: The mice presented significantly lower bone marrow adiposity (adipocyte volume/tissue volume, mean adipocyte number) but increased the bone parameters (bone mineral density, bone mineral content, bone volume/total volume) in the distal femoral metaphysis. CONCLUSION: Endogenous n-3 PUFAs protect bone marrow adipogenesis, which provides a novel drug target.


Assuntos
Adipogenia , Medula Óssea/metabolismo , Caderinas/fisiologia , Ácidos Graxos Ômega-3/fisiologia , Osteoporose/prevenção & controle , Ovariectomia , Adiposidade , Animais , Caderinas/genética , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , PPAR gama/análise
12.
Diabetes ; 59(2): 471-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19933995

RESUMO

OBJECTIVE: To evaluate the direct impact of n-3 polyunsaturated fatty acids (n-3 PUFAs) on the functions and viability of pancreatic beta-cells. RESEARCH DESIGN AND METHODS: We developed an mfat-1 transgenic mouse model in which endogenous production of n-3 PUFAs was achieved through overexpressing a C. elegans n-3 fatty acid desaturase gene, mfat-1. The islets and INS-1 cells expressing mfat-1 were analyzed for insulin secretion and viability in response to cytokine treatment. RESULTS: The transgenic islets contained much higher levels of n-3 PUFAs and lower levels of n-6 PUFAs than the wild type. Insulin secretion stimulated by glucose, amino acids, and glucagon-like peptide-1 (GLP-1) was significantly elevated in the transgenic islets. When challenged with tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and gamma-interferon (IFN-gamma), the transgenic islets completely resisted cytokine-induced cell death. Adenoviral transduction of mfat-1 gene in wild-type islets and in INS-1 cells led to acute changes in the cellular levels of n-3- and n-6 PUFAs and recapitulated the results in the transgenic islets. The expression of mfat-1 led to decreased production of prostaglandin E(2) (PGE(2)), which in turn contributed to the elevation of insulin secretion. We further found that cytokine-induced activation of NF-kappaB and extracellular signal-related kinase 1/2 (ERK(1/2)) was significantly attenuated and that the expression of pancreatic duodenal hemeobox-1 (PDX-1), glucokinase, and insulin-1 was increased as a result of n-3 PUFA production. CONCLUSIONS: Stable cellular production of n-3 PUFAs via mfat-1 can enhance insulin secretion and confers strong resistance to cytokine-induced beta-cell destruction. The utility of mfat-1 gene in deterring type 1 diabetes should be further explored in vivo.


Assuntos
Citocinas/farmacologia , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-6/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Morte Celular/efeitos dos fármacos , Primers do DNA , Sondas de DNA , DNA Complementar/genética , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Ômega-3/biossíntese , Vetores Genéticos , Glutamina/farmacologia , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Leucina/farmacologia , Camundongos , Camundongos Transgênicos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo
13.
Mol Cancer Ther ; 7(10): 3203-11, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18852124

RESUMO

Epidemiologic studies inclusively indicate that "unhealthy" dietary fat intake is one of the potential risk factors for cancer. In dietary fat, there are two types of polyunsaturated fatty acids (PUFA), omega-3 (n-3) and omega-6 (n-6). Numerous studies support that the ratio of n-6/n-3 affects tumorigenesis. It was reported that adenoviral transfer of the fat-1 gene, which converts n-6 to n-3, into breast and lung cancer cells had an antitumor effect in vitro. However, the effects of the fat-1 gene expression on tumor growth in vivo have not been studied and the mechanisms remain unclear. Accordingly, prostate cancer DU145 and PC3 cells were transfected with either the fat-1 gene or a control vector. The cells that expressed the fat-1 gene had a lower n-6/n-3 PUFA ratio compared with the cells that expressed the control vector. The fat-1 gene expression significantly inhibited prostate cancer cell proliferation and invasion in vitro. The fat-1 and control vector-transfected prostate cancer cells were s.c. implanted into severe combined immunodeficient mice for 6 weeks. The fat-1 gene expression significantly diminished tumor growth in vivo, but the control vector had no effect. Finally, we evaluated signaling pathways that may be important for fat-1 gene function. Administration of n-3 PUFA induced caspase-3-mediated prostate cancer cell apoptosis in vitro. The fat-1 gene expression inhibited prostate cancer cell proliferation via reduction of GSK-3beta phosphorylation and subsequent down-regulation of both beta-catenin and cyclin D1. These results suggest that fat-1 gene transfer directly into tumor cells could be used as a novel therapeutic approach.


Assuntos
Apoptose , Proteínas de Caenorhabditis elegans/genética , Ácidos Graxos Dessaturases/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Animais , Apoptose/efeitos dos fármacos , Caenorhabditis elegans , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-6/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Camundongos , Invasividade Neoplásica , Fosforilação/efeitos dos fármacos , Transfecção , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA