Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Orthop Surg Res ; 19(1): 127, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38326818

RESUMO

BACKGROUND: Icariin, a traditional Chinese medicine, has demonstrated anti-osteoporotic properties in ovariectomized mice. However, its effectiveness in preventing bone loss induced by ketogenic diet (KD), which mimics osteoporosis in human, remains unexplored. This study aims to investigate icariin's impact on KD-induced bone loss in mice. METHODS: Thirty mice were divided into: sham, KD, and KD + icariin groups. Post a 12-week intervention, evaluation including bone microstructures, serum concentrations of tartrate-resistant acid phosphatase (TRAP) and bone-specific alkaline phosphatase (ALP), and femoral tissue expression levels of osteocalcin (OCN) and TRAP. The expression levels of mammalian target of rapamycin (mTOR), ALP, peroxisome proliferator-activated receptor gamma (PPAR-γ), phosphorylated mTOR (p-mTOR), and the autophagy adaptor protein (p62) were also analyzed. Alizarin granule deposition and cellular ALP levels were measured following the induction of bone marrow mesenchymal stem cells (BMSCs) into osteogenesis. RESULTS: The study found that KD significantly impaired BMSCs' osteogenic differentiation, leading to bone loss. Icariin notably increased bone mass, stimulated osteogenesis, and reduced cancellous bone loss. In the KD + icariin group, measures such as bone tissue density (TMD), bone volume fraction (BV/TV), trabecular number (Tb.N), and trabecular thickness (Tb.Th) were significantly higher than in the KD group. Additionally, bone trabecular separation (Tb.Sp) was markedly lower in the KD + icariin group. Moreover, icariin increased OCN and ALP levels while suppressing PPAR-γ, TRAP, p62, and p-mTOR. In cellular studies, icariin encouraged osteogenic development in BMSCs under KD conditions. CONCLUSIONS: Icariin effectively counteracts bone thinning and improves bone microstructure. Its mechanism likely involves stimulating BMSCs osteogenic differentiation and inhibiting bone resorption, potentially through mTOR downregulation. These findings suggest icariin's potential as an alternative treatment for KD-induced bone loss.


Assuntos
Doenças Ósseas Metabólicas , Dieta Cetogênica , Flavonoides , Células-Tronco Mesenquimais , Osteoporose , Humanos , Camundongos , Animais , Osteogênese , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Osteoporose/tratamento farmacológico , Osteoporose/etiologia , Osteoporose/metabolismo , Diferenciação Celular , Doenças Ósseas Metabólicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Autofagia , Células-Tronco Mesenquimais/metabolismo , Células da Medula Óssea/metabolismo , Células Cultivadas , Mamíferos
2.
J Orthop Surg Res ; 19(1): 109, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38308345

RESUMO

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease caused by the deterioration of cartilage. However, the underlying mechanisms of OA pathogenesis remain elusive. METHODS: Hub genes were screened by bioinformatics analysis based on the GSE114007 and GSE169077 datasets. The Sprague-Dawley (SD) rat model of OA was constructed by intra-articular injection of a mixture of papain and L-cysteine. Hematoxylin-eosin (HE) staining was used to detect pathological changes in OA rat models. Inflammatory cytokine levels in serum were measured employing the enzyme-linked immunosorbent assay (ELISA). The reverse transcription quantitative PCR (RT-qPCR) was implemented to assess the hub gene expressions in OA rat models. The roles of PDK4 and the mechanism regulating the PPAR pathway were evaluated through western blot, cell counting kit-8 (CCK-8), ELISA, and flow cytometry assays in C28/I2 chondrocytes induced by IL-1ß. RESULTS: Six hub genes were identified, of which COL1A1, POSTN, FAP, and CDH11 expressions were elevated, while PDK4 and ANGPTL4 were reduced in OA. Overexpression of PDK4 inhibited apoptosis, inflammatory cytokine levels (TNF-α, IL-8, and IL-6), and extracellular matrix (ECM) degradation protein expressions (MMP-3, MMP-13, and ADAMTS-4) in IL-1ß-induced chondrocytes. Further investigation revealed that PDK4 promoted the expression of PPAR signaling pathway-related proteins: PPARA, PPARD, and ACSL1. Additionally, GW9662, an inhibitor of the PPAR pathway, significantly counteracted the inhibitory effect of PDK4 overexpression on IL-1ß-induced chondrocytes. CONCLUSION: PDK4 inhibits OA development by activating the PPAR pathway, which provides new insights into the OA management.


Assuntos
Osteoartrite , Receptores Ativados por Proliferador de Peroxissomo , Ratos , Animais , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Osteoartrite/metabolismo , Células Cultivadas , Ratos Sprague-Dawley , Condrócitos/metabolismo , Citocinas/metabolismo , Interleucina-1beta/metabolismo , Inflamação/metabolismo
3.
Exp Cell Res ; 434(1): 113878, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-38086504

RESUMO

Liver fibrosis is a significant health burden worldwide and has emerged as the leading cause of Hepatocellular carcinoma (HCC) incidence. Mitochondria are the dynamic organelles that regulate the differentiation, survival, and polarization of macrophages. Nuclear-DNA-associated proteins, micro-RNAs, as well as macrophage polarization are essential for maintaining intracellular and extra-cellular homeostasis in the liver parenchyma. Dysregulated mitochondrial coding genes (ETS complexes I, II, III, IV, and V), non-coding RNAs (mitomiRs), and nuclear alteration lead to the production of reactive oxygen species (ROS) and inflammation which are implicated in the transition of liver fibrosis into HCC. Recent findings indicated the protecting effect of E74-like factor 3/peroxisome proliferator-activated receptor-γ (Elf-3/PPAR-γ). HDAR-y inhibits the deacetylation of PPAR-y and maintains the PPAR-y pathway. Elf-3 plays a tumor suppressive role through epithelial-mesenchymal transition-related gene and zinc finger E-box binding homeobox 2 (ZEB-2) domain. Additionally, the development of HCC includes the PI3K/Akt/mTOR and transforming Growth Factor ß (TGF-ß) pathway that promotes the Epithelial-mesenchymal transition (EMT) through Smad/Snail/Slug signaling cascade. In contrast, the TLR2/NOX2/autophagy axis promotes M2 polarization in HCC. Thus, a thorough understanding of the mitochondrial and nuclear reciprocal relationship related to macrophage polarization could provide new research opportunities concerning diseases with a significant impact on liver parenchyma towards developing liver fibrosis or liver cancer. Moreover, this knowledge can be used to develop new therapeutic strategies to treat liver diseases.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Linhagem Celular Tumoral , Transdução de Sinais , Cirrose Hepática/patologia , Mitocôndrias/metabolismo , Macrófagos/metabolismo , Transição Epitelial-Mesenquimal
4.
Inflammopharmacology ; 32(1): 561-573, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37921960

RESUMO

Nitro-conjugated linoleic acid (NO2-CLA) has been observed to manifest salutary signaling responses, including anti-inflammatory and antioxidant properties. Here, the authors have explored the influence and underlying mechanisms of NO2-CLA on the proinflammatory reaction of murine macrophages that were challenged with lipopolysaccharide (LPS) derived from Prevotella intermedia, a putative periodontopathic bacterium. Treatment of LPS-activated RAW264.7 cells with NO2-CLA notably dampened the secretion of iNOS-derived NO, IL-1ß and IL-6 as well as their gene expressions and significantly enhanced the markers for M2 macrophage polarization. NO2-CLA promoted the HO-1 expression in cells challenged with LPS, and tin protoporphyrin IX, an HO-1 inhibitor, significantly reversed the NO2-CLA-mediated attenuation of NO secretion, but not IL-1ß or IL-6. We found that cells treated with NO2-CLA significantly increased mRNA expression of PPAR-γ compared to control cells, and NO2-CLA significantly reverted the decrease in PPAR-γ mRNA caused by LPS. Nonetheless, antagonists to PPAR-γ were unable to reverse the NO2-CLA-mediated suppression of inflammatory mediators. In addition, NO2-CLA did not alter the p38 and JNK activation elicited by LPS. Both NF-κB reporter activity and IκB-α degradation caused by LPS were notably diminished by NO2-CLA. NO2-CLA was observed to interrupt the nuclear translocation and DNA binding of p50 subunits caused by LPS with no obvious alterations in p65 subunits. Further, NO2-CLA attenuated the phosphorylation of STAT1/3 elicited in response to LPS. We propose that NO2-CLA could be considered as a possible strategy for the therapy of periodontal disease, although additional researches are certainly required to confirm this.


Assuntos
Ácidos Linoleicos Conjugados , Lipopolissacarídeos , Animais , Camundongos , Lipopolissacarídeos/farmacologia , Prevotella intermedia/química , Interleucina-6/metabolismo , Ácidos Linoleicos Conjugados/farmacologia , Ácidos Linoleicos Conjugados/metabolismo , Dióxido de Nitrogênio/metabolismo , Dióxido de Nitrogênio/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Macrófagos , RNA Mensageiro/metabolismo
5.
Fish Shellfish Immunol ; 145: 109302, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38128680

RESUMO

Feeding high-fat (HF) diets has been shown to cause hepatic and intestinal impairment in fish species, but the mode of action, especially the pathways involved in the intestine, has not been determined yet. In this study, the effects of resveratrol (RES) supplementation on the intestinal structure, microbial flora, and fat metabolism in red tilapia (Oreochromis niloticus) were determined. The results showed RES maintained the structural integrity of the intestine and significantly increased the number of goblet cells in the midgut. RES significantly induced interferon (IL)-1ß, IL-6, IL-10, and tumor necrosis factor (TNF)-α, serumal and fecal trimetlylamine oxide (TMAO) and lipopolysaccharides (LPS), intestinal acetic acid levels. However, the concentrations of bound bile acids increased in HF-fed red tilapia. Atp5fa1 and Pafah1b3 significantly increased, Pmt and Acss2 significantly decreased, respectively, with RES supplementation, which was alleviated and retained at the same level in the selisistat (EX527) group. While for transcriptome and proteomics results, RES was found to promote fatty acid ß-oxidation and arachidonic acid metabolism associated with the peroxisome proliferator-activated receptor (PPAR) signaling pathway. The next validation experiment showed some genes related to apoptosis and fatty acid metabolism pathways were altered by RES supplementation. Namely, sn6, loc100702698, new_14481, and prkaa1 were upregulated, while ffrs1, ap3s1, and loc100705861 were downregulated. RES significantly increased Planctomycetes and Verrucomicrobia while decreased Moonvirus, Citrobacter, and Pseudomonas. Akkermansia and Fusobacterium significantly increased and Aeromonas significantly decreased. Thus, unsaturated fatty acid biosynthesis significantly increased and carbohydrate/energy metabolism decreased. To conclude, RES enabled the body to complete fatty acid ß-oxidation and arachidonic acid metabolism, whereas the addition of inhibitors increased the expression of the phagosome transcriptome and reduced fatty acid ß-oxidative metabolism.


Assuntos
Ciclídeos , Tilápia , Animais , Tilápia/metabolismo , Ciclídeos/metabolismo , Dieta Hiperlipídica , Resveratrol/metabolismo , Metabolismo dos Lipídeos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Intestinos , Transdução de Sinais , Ácidos Graxos/metabolismo , Ácidos Araquidônicos/metabolismo , Ácidos Araquidônicos/farmacologia , Dieta , Suplementos Nutricionais , Ração Animal/análise
6.
Clin Res Hepatol Gastroenterol ; 47(10): 102231, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37865226

RESUMO

INTRODUCTION: Linalool is a monoterpene that occurs naturally in various aromatic plants and is identified in our previous study as a potential candidate for protection against high-fat diet (HFD)-induced metabolic dysfunction-associated steatotic liver disease (MASLD). However, little is known about its direct effects on hepatic lipid metabolism and oxidative stress. Therefore, this study aims to investigate the therapeutic effect of linalool against MASLD and the underlying mechanism. METHODS: To establish a rat model of MASLD, male Wistar rats were fed HFD for 16 weeks and orally administered linalool (100 mg/kg body weight) for 45 days starting from week 14. RESULTS: Linalool significantly reduced HFD-induced liver lipid accumulation and restored altered adipokine levels. Mechanistically, linalool downregulated the mRNA expression of sterol regulatory element binding protein 1 and its lipogenesis target genes fatty acid synthase and acetyl-CoA carboxylase, and upregulated the mRNA expression of genes involved in fatty acid oxidation (peroxisome proliferator-activated receptor (PPAR)-alpha [PPAR-α], lipoprotein lipase and protein kinase B [Akt]) as well as the upstream mediators sirtuin 1 (Sirt1) and AMP-activated protein kinase (AMPK) in the liver of MASLD rats. In addition, linalool also curbed oxidative stress by increasing antioxidant enzymes and activating nuclear erythroid-2-related factor 2 (Nrf-2) and its downstream target genes involved in antioxidant properties. CONCLUSION: Therefore, this study concludes that linalool attenuates lipid accumulation in the liver by inhibiting de novo lipogenesis, promoting fatty acid oxidation, and attenuating oxidative stress by regulating Sirt1/Akt/PPRA-α/AMPK and Nrf-2/ HO-1 signaling pathways.


Assuntos
Fígado Gorduroso , Hepatopatia Gordurosa não Alcoólica , Ratos , Animais , Sirtuína 1/metabolismo , Sirtuína 1/farmacologia , Sirtuína 1/uso terapêutico , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases Ativadas por AMP/metabolismo , Antioxidantes/uso terapêutico , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/uso terapêutico , Ratos Wistar , Fígado/metabolismo , Metabolismo dos Lipídeos , Transdução de Sinais , Estresse Oxidativo , Ácidos Graxos , Lipídeos , RNA Mensageiro/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico
7.
Int J Biol Macromol ; 247: 125745, 2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37423454

RESUMO

P-glycoprotein (Pgp), a member of ATP binding cassette (ABC) transporter family, can extrude toxic substances out of cells by mediating multi-xenobiotic resistance (MXR) in aquatic organisms, however, its regulation and association with MXR are still unclear. In this work, the genetic information of Pgp in freshwater crab Sinopotamon henanense (ShPgp) was revealed for the first time. ShPgp with a total of 4488 bp was cloned and analyzed, which includes 4044 bp open reading frame, 353 bp 3' untranslated region, and 91 bp 5' untranslated region. The recombinant ShPGP were expressed in Saccharomyces cerevisiae and taken for SDS-PAGE and western blot analysis. ShPGP was widely expressed in the midgut, hepatopancreas, testis, ovary, gill, hemocytes, accessory gonad and myocardium of the crabs studied. The images of immunohistochemistry indicated that ShPgp was mainly distributed in the cytoplasm and cell membrane. When the crabs were exposed to cadmium or cadmium containing quantum dots (Cd-QDs), not only the relative expression of ShPgp mRNA and the protein produced were enhanced, but also the MXR activity and ATP contents. The relative expression of target genes related to energy metabolism, detoxification and apoptosis was also determined in the carbs exposed to Cd or Cd-QDs. The results showed that bcl-2 was significantly down-regulated, while other genes were up-regulated except PPAR (not affected). However, when the Shpgp in treated crabs was interfering by knockdown technique, their apoptosis and the expression of proteolytic enzyme genes and transcription factors MTF1 and HSF1 were also elevated, while the expression of apoptosis inhibiting and fat metabolism genes were compromised. Based on the observation, we concluded that MTF1 and HSF1 were involved in gene transcription regulation of mt and MXR, respectively, while PPAR had limited regulatory effect on those genes in S. henanense. NF-κB may play a negligible role in the process of apoptosis in testes induced by cadmium or Cd-QDs. However, the detail information regarding Pgp involvement in SOD or MT, and its association with apoptosis during xenobiotics insults remain to be explored.


Assuntos
Braquiúros , Pontos Quânticos , Poluentes Químicos da Água , Animais , Feminino , Braquiúros/genética , Braquiúros/metabolismo , Cádmio/metabolismo , Distribuição Tecidual , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Glicoproteínas/metabolismo , Água Doce , Clonagem Molecular , Trifosfato de Adenosina/metabolismo , Poluentes Químicos da Água/metabolismo
8.
Cells ; 12(12)2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37371027

RESUMO

Sulfite predominantly accumulates in the brain of patients with isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies. Patients present with severe neurological symptoms and basal ganglia alterations, the pathophysiology of which is not fully established. Therapies are ineffective. To elucidate the pathomechanisms of ISOD and MoCD, we investigated the effects of intrastriatal administration of sulfite on myelin structure, neuroinflammation, and oxidative stress in rat striatum. Sulfite administration decreased FluoromyelinTM and myelin basic protein staining, suggesting myelin abnormalities. Sulfite also increased the staining of NG2, a protein marker of oligodendrocyte progenitor cells. In line with this, sulfite also reduced the viability of MO3.13 cells, which express oligodendroglial markers. Furthermore, sulfite altered the expression of interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interleukin-10 (IL-10), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and heme oxygenase-1 (HO-1), indicating neuroinflammation and redox homeostasis disturbances. Iba1 staining, another marker of neuroinflammation, was also increased by sulfite. These data suggest that myelin changes and neuroinflammation induced by sulfite contribute to the pathophysiology of ISOD and MoCD. Notably, post-treatment with bezafibrate (BEZ), a pan-PPAR agonist, mitigated alterations in myelin markers and Iba1 staining, and IL-1ß, IL-6, iNOS and HO-1 expression in the striatum. MO3.13 cell viability decrease was further prevented. Moreover, pre-treatment with BEZ also attenuated some effects. These findings show the modulation of PPAR as a potential opportunity for therapeutic intervention in these disorders.


Assuntos
Bezafibrato , Receptores Ativados por Proliferador de Peroxissomo , Ratos , Animais , Bezafibrato/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Bainha de Mielina , Doenças Neuroinflamatórias , Interleucina-6/farmacologia , Estresse Oxidativo , Sulfitos/farmacologia
9.
Can J Physiol Pharmacol ; 101(8): 382-392, 2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37224567

RESUMO

This study aims to evaluate the effect of diclofenac addition to the preservation solution Celsior on liver graft preservation. Liver from Wistar rats were cold flushed in situ, harvested, and then stored in Celsior solution (24 h, 4 °C) supplemented or not with 50 mg/L of diclofenac sodium salt. Reperfusion was performed (120 min, 37 °C) using the isolated perfusion rat liver model. Perfusate samples were collected to evaluate transaminases' activities after cold storage and by the end of reperfusion. To evaluate liver function, bile flow, hepatic clearance of bromosulfophthalein, and vascular resistance were assessed. Diclofenac scavenging property (DPPH assay) as well as oxidative stress parameters (SOD and MPO activities and the concentration of glutathione, conjugated dienes, MDA, and carbonylated proteins) were measured. Transcription factors (PPAR-γ and NF-κB), inflammation (COX-2, IL-6, HMGB-1, and TLR-4), as well as apoptosis markers (Bcl-2 and Bax) were determined by quantitative RT-PCR. Enriching the preservation solution Celsior with diclofenac sodium salt attenuated liver injuries and improved graft function. Oxidative stress, inflammation, and apoptosis were significantly reduced in Celsior + Diclo solution. Also, diclofenac activated PPAR-γ and inhibited NF-κB transcription factors. To decrease graft damage and improve transplant recovery, diclofenac sodium salt may be a promising additive to preservation solution.


Assuntos
Soluções para Preservação de Órgãos , Traumatismo por Reperfusão , Ratos , Animais , Diclofenaco/farmacologia , Soluções para Preservação de Órgãos/farmacologia , Soluções para Preservação de Órgãos/metabolismo , NF-kappa B/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Ratos Wistar , Fígado , Glutationa/metabolismo , Inflamação/metabolismo , Traumatismo por Reperfusão/metabolismo , Preservação de Órgãos
10.
Int J Biol Macromol ; 241: 124654, 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37119902

RESUMO

MicroRNAs (miRNAs) are class of 22 nt short RNA sequences which inhibit protein translation through binding to the 3'UTR of its target genes. The continuous ovulatory property of chicken follicle makes it a perfect model for studying granulosa cell (GC) functions. In this study, we found that large number of miRNAs including miR-128-3p, were differentially expressed in the GCs of F1 and F5 follicles of chicken. Subsequently, the results revealed that miR-128-3p inhibited proliferation, the formation of lipid droplets, and hormone secretion in chicken primary GCs through directly targeting YWHAB and PPAR-γ genes. To determine the effects of 14-3-3ß (encoded by YWHAB) protein on GCs functions, we overexpressed or inhibited the expression of YWHAB, and the results showed that YWHAB inhibited the function of FoxO proteins. Collectively, we found that miR-128-3p was highly expressed in the chicken F1 follicles compared to the F5 follicles. In addition, the results indicated that miR-128-3p promoted GC apoptosis through 14-3-3ß/FoxO pathway via repressing YWHAB, and inhibited lipid synthesis by impeding the PPAR-γ/LPL pathway, as well as reduced the secretion of progesterone and estrogen. Taken together, the results showed that miR-128-3p plays a regulatory role in chicken granulosa cell function via 14-3-3ß/FoxO and PPAR-γ/LPL signaling pathways.


Assuntos
Galinhas , MicroRNAs , Animais , Feminino , Galinhas/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Células da Granulosa/metabolismo , Transdução de Sinais , Proliferação de Células/genética
11.
Fundam Clin Pharmacol ; 37(5): 947-959, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36977287

RESUMO

Adenosine monophosphate kinase/liver kinase B1/peroxisome proliferator-activated receptor-γ coactivator 1-α (AMPK/LKB1/PGC1α) pathway has a vital role in regulating age-related diseases. It controls neurogenesis, cell proliferation, axon outgrowth, and cellular energy homeostasis. AMPK pathway also regulates mitochondrial synthesis. The current study evaluated the effect of chrysin on D-galactose (D-gal) induced-aging, neuron degeneration, mitochondrial dysfunction, oxidative stress, and neuroinflammation in mice. The mice were allocated randomly into four groups (10 each group): Group 1: normal control group, Group 2: D-gal group, Groups 3 and 4: chrysin (125 and 250 mg/kg, respectively). Groups 2-4 were injected with D-gal (200 mg/kg/day; s.c) for 8 weeks to induce aging. Groups 3 and 4 were orally gavaged every day concurrent with D-gal. At the end of experiment, behavioral, brain biochemical and histopathological changes were monitored. Chrysin administration elevated discrimination ratio in object recognition, Y Maze percentage alternation, locomotor activity and brain contents of AMPK, LKB1, PGC1α, NAD (P)H quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO-1), nerve growth factor (NGF) (neurotrophin-3; NT-3), and seretonin as well as reduced brain contents of tumor necrosis factor-alpha (TNF-α), nuclear factor kappa B (NF-κB), advanced glycation end products (AGEs) and glial fibrillary acidic protein (GFAP) compared to D-gal-treated mice. Chrysin also alleviated cerebral cortex and white matter neurons degeneration. Chrysin protects against neurodegeneration, improves mitochondrial autophagy and biogenesis as well as activates antioxidant genes expression. In addition, chrysin ameliorates neuroinflammation and stimulates the release of NGF and serotonin neurotransmitter. So, chrysin has a neuroprotective effect in D-gal induced-aging in mice.


Assuntos
Adenilato Quinase , Galactose , Camundongos , Animais , Galactose/farmacologia , Adenilato Quinase/metabolismo , Adenilato Quinase/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Regulação para Cima , Proteínas Quinases Ativadas por AMP/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/farmacologia , Doenças Neuroinflamatórias , Fator de Crescimento Neural/metabolismo , Transdução de Sinais , Envelhecimento , Estresse Oxidativo , Fígado/metabolismo
12.
J Biochem Mol Toxicol ; 37(6): e23350, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36988379

RESUMO

Doxorubicin (DOX) is one of the basic anticancer drugs, nonetheless its use is restricted due to noxious side effects. Kidney failure is one of the main side effects that restrict its medical use. The current study assessed the nephroprotective effects of fenofibrate and pioglitazone against the renal injury induced by doxorubicin in rats and illustrated the probable mechanisms underlying these protective effects. For this purpose, Male Sprague-Dawley rats weighing (200-230 g) were allocated into seven groups treated for 15 days as following: control (50% corn oil + 50% DMSO p.o), fenofibrate (100 mg/kg p.o) and pioglitazone (10 mg/kg p.o) as well as four groups of DOX (15 mg/kg i.p on 11th day). DOX groups included DOX alone and DOX with protective drugs fenofibrate, pioglitazone or both of them. As a result of doxorubicin nephrotoxicity; serum creatinine and blood urea nitrogen were remarkably elevated. Moreover, renal glutathione was significantly reduced while tissue lipid peroxidation malondialdehyde, tumor necrosis factor-α, nuclear factor-kappa B p65 (NF-κB p65), interleukin-1ß, p38 mitogen activated protein kinase (p38-MAPK) and caspase-3 (Casp-3) were significantly augmented. Treatment with fenofibrate and pioglitazone either alone or in combination markedly attenuated DOX-induced injury by suppression of oxidative stress, inflammation and apoptosis. The above-mentioned biochemical markers were affirmed by histological assessment. In conclusion, fenofibrate, pioglitazone, and their combination possess potential prophylactic effects against doxorubicin-induced renal injury through modulation of p38-MAPK/NF-κB p65 pathway with superiority to the combination.


Assuntos
Fenofibrato , Insuficiência Renal , Ratos , Masculino , Animais , NF-kappa B/metabolismo , Ratos Sprague-Dawley , Pioglitazona/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Fenofibrato/farmacologia , Fenofibrato/metabolismo , Rim , Doxorrubicina/efeitos adversos , Estresse Oxidativo , Hipoglicemiantes/farmacologia , Apoptose
13.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(1): 154-161, 2023 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-36765493

RESUMO

OBJECTIVE: To investigate the effect of adipocytes in the bone marrow microenvironment of patients with multiple myeloma (MM) on the pathogenesis of MM. METHODS: Bone marrow adipocytes (BMA) in bone marrow smears of health donors (HD) and newly diagnosed MM (ND-MM) patients were evaluated with oil red O staining. The mesenchymal stem cells (MSC) from HD and ND-MM patients were isolated, and in vitro co-culture assay was used to explore the effects of MM cells on the adipogenic differentiation of MSC and the role of BMA in the survival and drug resistance of MM cells. The expression of adipogenic/osteogenic differentiation-related genes PPAR-γ, DLK1, DGAT1, FABP4, FASN and ALP both in MSC and MSC-derived adipocytes was determined with real-time quantitative PCR. The Western blot was employed to detect the expression levels of IL-6, IL-10, SDF-1α, TNF-α and IGF-1 in the supernatant with or without PPAR-γ inhibitor. RESULTS: The results of oil red O staining of bone marrow smears showed that BMA increased significantly in patients of ND-MM compared with the normal control group, and the BMA content was related to the disease status. The content of BMA decreased in the patients with effective chemotherapy. MM cells up-regulated the expression of MSC adipogenic differentiation-related genes PPAR-γ, DLK1, DGAT1, FABP4 and FASN, but the expression of osteogenic differentiation-related gene ALP was significantly down-regulated. This means that the direct consequence of the interaction between MM cells and MSC in the bone marrow microenvironment is to promote the differentiation of MSC into adipocytes at the expense of osteoblasts, and the cytokines detected in supernatant changed. PPAR-γ inhibitor G3335 could partially reverse the release of cytokines by BMA. Those results confirmed that BMA regulated the release of cytokines via PPAR-γ signal, and PPAR-γ inhibitor G3335 could distort PPAR-γ mediated BMA maturation and cytokines release. The increased BMA and related cytokines effectively promoted the proliferation, migration and drug resistance of MM cells. CONCLUSION: The BMA and its associated cytokines are the promoting factors in the survival, proliferation and migration of MM cells. BMA can protect MM cells from drug-induced apoptosis and plays an important role in MM treatment failure and disease progression.


Assuntos
Mieloma Múltiplo , Osteogênese , Humanos , Osteogênese/genética , Medula Óssea/metabolismo , Mieloma Múltiplo/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Diferenciação Celular , Adipogenia , Citocinas/metabolismo , Adipócitos/metabolismo , Células da Medula Óssea/metabolismo , Células Cultivadas , PPAR gama/metabolismo , PPAR gama/farmacologia , Microambiente Tumoral
14.
Nutr Res ; 110: 74-86, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36689814

RESUMO

Obesity is a chronic metabolic disease that involves excessive accumulation of fat in white adipose tissue (WAT). Apart from storing excess fats, WAT also serves as an important endocrine organ secreting adipocytokines such as adiponectin and leptin. Adiponectin and leptin bind to their transmembrane receptors adiponectin receptor 1 (AdipoR1)/adiponectin receptor 2 (AdipoR2) and Ob-R, respectively, and mediate their effect on metabolism by regulating multiple downstream targets. Dietary fat is considered the main culprit behind obesity development. Numerous preclinical studies have highlighted role of essential polyunsaturated fatty acids (PUFAs), particularly n-3 PUFAs, in prevention of obesity. Despite emerging data, there still is no clear understanding of the mechanism of action of n-3 PUFAs and n-6 PUFAs on adipose tissue function in two functionally and anatomically different depots of WAT: visceral and subcutaneous. We designed this study using a high fat diet (HFD) fed rodent model of obesity to test our hypothesis that n-3 and n-6 PUFAs possibly differentially modulate adipokine secretion and downstream metabolic pathways such as peroxisome proliferator-activated receptor-γ (PPAR-γ), protein kinase B (AKT)-forkhead box O1 (FOXO1), and Janus kinase-signal transducer and activator of transcription in obesity. The results of the current study showed that n-3 PUFAs upregulate the expression of AdipoR1/R2 and ameliorate the effects of HFD by modulating adipogenesis via PPAR-γ and by improving glucose tolerance and lipid metabolism via AKT-FOXO1 axis in fish oil fed rats. However, n-6 PUFAs did not show any remarkable change compared with HFD fed animals. Our study highlights that n-3 PUFAs modulate expression of various targets in adiponectin and leptin signaling cascade, bringing about an overall reduction in obesity and improvement in adipose tissue function in HFD induced obesity.


Assuntos
Dieta Hiperlipídica , Ácidos Graxos Ômega-3 , Ratos , Animais , Dieta Hiperlipídica/efeitos adversos , Adiponectina , Leptina/metabolismo , Ratos Wistar , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácidos Graxos Ômega-6/farmacologia , Receptores de Adiponectina/metabolismo , Receptores de Adiponectina/uso terapêutico , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/uso terapêutico , Obesidade/metabolismo , Tecido Adiposo Branco/metabolismo , Tecido Adiposo/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Ácidos Graxos Ômega-3/uso terapêutico , Adipocinas/metabolismo , Ácidos Graxos Insaturados/metabolismo , Transdução de Sinais
15.
Plast Reconstr Surg ; 151(5): 1005-1015, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36534068

RESUMO

BACKGROUND: The pathophysiology of adipose proliferation or differentiation in extremity lymphedema has not been thoroughly studied. This study investigated the impacts of the lymph harvested from lymphedematous limbs on the adipogenesis of adipose-derived stem cells (ASCs). METHODS: ASCs were isolated from the adipose tissue of normal extremities and cultured with lymph collected from Cheng lymphedema grade III to IV patients or adipogenic differentiation medium (ADM) and further subjected to differentiation and proliferation assay. The expression of adipogenesis genes was examined by real-time polymerase chain reaction to investigate the effect of lymph on ASCs. The level of adipogenic cytokines in the lymph was also evaluated. RESULTS: The adipocytes were significantly larger in lymphedema fat tissue compared with that in normal fat tissues ( P < 0.00). The adipogenesis of ASCs cultured in lymph was significantly enhanced compared with in ADM ( P = 0.008) on day 10, suggesting that the adipogenesis of ASCs was promoted under the lymph-cultured environment. The expression of adipogenesis genes, peroxisome proliferator-activated receptor ( P = 0.02), CAAT/enhancer-binding protein α ( P = 0.008); fatty-acid binding protein ( P = 0.004), and lipoprotein lipase ( P = 0.003), was statistically elevated when the ASCs were cultured with lymph. The insulin content in lymph was statistically higher in lymph ( P < 0.001) than in plasma. CONCLUSIONS: The adipogenesis of ASCs was promoted under the lymph-cultured environment with statistically increased adipogenesis genes of peroxisome proliferator-activated receptor, CAAT/enhancer-binding protein α, fatty-acid binding protein, and lipoprotein lipase. The excess lymph accumulated in the lymphedematous extremity contained a greater insulin/insulin-like growth factor-2. These adipogenic factors promoted the expression of early adipogenesis genes and led ASCs to undergo adipogenesis and differentiated into adipocytes. CLINICAL RELEVANCE STATEMENT: The accumulation of adipose tissue in the lymphedema region was contributed from the content of excess lymph.


Assuntos
Insulinas , Linfedema , Humanos , Adipogenia/fisiologia , Lipase Lipoproteica/metabolismo , Lipase Lipoproteica/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Adipócitos/fisiologia , Tecido Adiposo , Diferenciação Celular/genética , Células-Tronco/fisiologia , Insulinas/metabolismo , Insulinas/farmacologia , Células Cultivadas
16.
Nutr Res ; 106: 35-46, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36126528

RESUMO

The hypothesis of the present study was that nitro-fatty acids (NO2-FAs) would suppress inflammation associated with periodontal disease. To test this hypothesis, we investigated the influence of nitrooleic acid, a prototypical NO2-FA, on the inflammatory response of murine macrophages activated with lipopolysaccharide (LPS) from Prevotella intermedia, a pathogen associated the etiology of different types of periodontal diseases. LPS was prepared from P. intermedia cells by using phenol-water protocol. Culture supernatants were assayed for nitric oxide (NO), interleukin-1ß (IL-1ß), and IL-6. Real-time polymerase chain reaction and immunoblotting analyses were performed to quantify messenger RNA and protein expression, respectively. The secreted embryonic alkaline phosphatase reporter assay was performed to measure NF-κB activation. The transcription factor assay kit was used to measure DNA-binding of NF-κB subunits. Findings obtained from the present study revealed that nitrooleic acid suppresses the generation and messenger RNA expression of inducible NO synthase-derived NO, IL-1ß, and IL-6 in RAW264.7 cells activated with P. intermedia LPS and promotes macrophage polarization toward anti-inflammatory M2 phenotype. We also found that nitrooleic acid exerts its effect via heme oxygenase-1 induction and suppression of NF-κB signaling. The inhibition of NO and proinflammatory cytokine production by nitrooleic acid was independent from PPAR-γ, JNK, p38, and STAT1/3. Nitrooleic acid may represent a novel class of agent as a host modulator which has therapeutic benefit in periodontal disease, though more work is needed to confirm this.


Assuntos
Lipopolissacarídeos , Doenças Periodontais , Fosfatase Alcalina/metabolismo , Animais , Anti-Inflamatórios/farmacologia , DNA , Ácidos Graxos/farmacologia , Heme Oxigenase-1/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Camundongos , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Dióxido de Nitrogênio/metabolismo , Dióxido de Nitrogênio/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Fenóis/farmacologia , Prevotella intermedia/genética , Prevotella intermedia/metabolismo , RNA Mensageiro , Água/metabolismo , Água/farmacologia
17.
Phytother Res ; 36(10): 3859-3884, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35989419

RESUMO

Crocin is a hydrophilic carotenoid that is synthesized in the flowers of the Crocus genus. Numerous in vitro and in vivo research projects have been published about the biological and pharmacological properties and toxicity of crocin. Crocin acts as a memory enhancer, anxiolytic, aphrodisiac, antidepressant, neuroprotective, and so on. Here, we introduce an updated and comprehensive review of crocin molecular mechanisms based on previously examined and mentioned in the literature. Different studies confirmed the significant effect of crocin to control pathological conditions, including oxidative stress, inflammation, metabolic disorders, neurodegenerative disorders, and cancer. The neuroprotective effect of crocin could be related to the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT)/mammalian target of rapamycin (mTOR), Notch, and cyclic-AMP response element-binding protein signaling pathways. The crocin also protects the cardiovascular system through the inhibitory effect on toll-like receptors. The regulatory effect of crocin on PI3K/AKT/mTOR, AMP-activated protein kinase, mitogen-activated protein kinases (MAPK), and peroxisome proliferator-activated receptor pathways can play an effective role in the treatment of metabolic disorders. The crocin has anticancer activity through the PI3K/AKT/mTOR, MAPK, vascular endothelial growth factor, Wnt/ß-catenin, and Janus kinases-signal transducer and activator of transcription suppression. Also, the nuclear factor-erythroid factor 2-related factor 2 and p53 signaling pathway activation may be effective in the anticancer effect of crocin. Finally, among signaling pathways regulated by crocin, the most important ones seem to be those related to the regulatory effect on the PI3K/AKT/mTOR pathway.


Assuntos
Ansiolíticos , Afrodisíacos , Fármacos Neuroprotetores , Monofosfato de Adenosina/farmacologia , Ansiolíticos/farmacologia , Afrodisíacos/farmacologia , Carotenoides/farmacologia , Carotenoides/uso terapêutico , Janus Quinases/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fármacos Neuroprotetores/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo
18.
Cell Stress Chaperones ; 27(5): 499-511, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35779187

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a major health concern. Endoplasmic reticulum (ER) stress, inflammation, and metabolic dysfunctions may be targeted to prevent the progress of nonalcoholic fatty liver disease. Sulforaphane (SFN), a sulfur-containing compound that is abundant in broccoli florets, seeds, and sprouts, has been reported to have beneficial effects on attenuating metabolic diseases. In light of this, the present study was designed to elucidate the mechanisms by which SFN ameliorated ER stress, inflammation, lipid metabolism, and insulin resistance - induced by a high-fat diet and ionizing radiation (IR) in rats. In our study, the rats were randomly divided into five groups: control, HFD, HFD + SFN, HFD + IR, and HFD + IR + SFN groups. After the last administration of SFN, liver and blood samples were taken. As a result, the lipid profile, liver enzymes, glucose, insulin, IL-1ß, adipokines (leptin and resistin), and PI3K/AKT protein levels, as well as the mRNA gene expression of ER stress markers (IRE-1, sXBP-1, PERK, ATF4, and CHOP), fatty acid synthase (FAS), peroxisome proliferator-activated receptor-α (PPAR-α). Interestingly, SFN treatment modulated the levels of proinflammatory cytokine including IL-1ß, metabolic indices (lipid profile, glucose, insulin, and adipokines), and ER stress markers in HFD and HFD + IR groups. SFN also increases the expression of PPAR-α and AMPK genes in the livers of HFD and HFD + IR groups. Meanwhile, the gene expression of FAS and CHOP was significantly attenuated in the SFN-treated groups. Our results clearly show that SFN inhibits liver toxicity induced by HFD and IR by ameliorating the ER stress events in the liver tissue through the upregulation of AMPK and PPAR-α accompanied by downregulation of FAS and CHOP gene expression.


Assuntos
Estresse do Retículo Endoplasmático , Insulinas , Isotiocianatos , Hepatopatia Gordurosa não Alcoólica , Sulfóxidos , Proteínas Quinases Ativadas por AMP/genética , Animais , Citocinas/metabolismo , Dieta Hiperlipídica , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Ácido Graxo Sintases/farmacologia , Glucose/metabolismo , Insulinas/genética , Insulinas/metabolismo , Insulinas/farmacologia , Isotiocianatos/farmacologia , Isotiocianatos/uso terapêutico , Leptina , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/genética , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Ratos , Resistina/genética , Resistina/metabolismo , Resistina/farmacologia , Sulfóxidos/farmacologia , Sulfóxidos/uso terapêutico , Regulação para Cima
19.
J Cachexia Sarcopenia Muscle ; 13(5): 2480-2491, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35903870

RESUMO

BACKGROUND: Chemotherapy induces a cachectic-like phenotype, accompanied by skeletal muscle wasting, weakness and mitochondrial dysfunction. Peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC1α), a regulator of mitochondrial biogenesis, is often reduced in cachectic skeletal muscle. Overexpression of PGC1α has yielded mixed beneficial results in cancer cachexia, yet investigations using such approach in a chemotherapy setting are limited. Utilizing transgenic mice, we assessed whether overexpression of PGC1α could combat the skeletal muscle consequences of cisplatin. METHODS: Young (2 month) and old (18 month) wild-type (WT) and PGC1α transgenic male and female mice (Tg) were injected with cisplatin (C; 2.5 mg/kg) for 2 weeks, while control animals received saline (n = 5-9/group). Animals were assessed for muscle mass and force, motor unit connectivity, and expression of mitochondrial proteins. RESULTS: Young WT + C mice displayed reduced gastrocnemius mass (male: -16%, P < 0.0001; female: -11%, P < 0.001), muscle force (-6%, P < 0.05, both sexes), and motor unit number estimation (MUNE; male: -53%, P < 0.01; female: -51%, P < 0.01). Old WT + C male and female mice exhibited gastrocnemius wasting (male: -22%, P < 0.05; female: -27%, P < 0.05), muscle weakness (male: -20%, P < 0.0001; female: -17%, P < 0.01), and loss of MUNE (male: -82%, P < 0.01; female: -62%, P < 0.05), suggesting exacerbated cachexia compared with younger animals. Overexpression of PGC1α had mild protective effects on muscle mass in young Tg + C male only (gastrocnemius: +10%, P < 0.05); however, force and MUNE were unchanged in both young Tg + C male and female, suggesting preservation of neuromuscular function. In older male, protective effects associated with PGC1α overexpression were heighted with Tg + C demonstrating preserved muscle mass (gastrocnemius: +34%, P < 0.001), muscle force (+13%, P < 0.01), and MUNE (+3-fold, P < 0.05). Similarly, old female Tg + C did not exhibit muscle wasting or reductions in MUNE, and had preserved muscle force (+11%, P < 0.05) compared with female WT + C. Follow-up molecular analysis demonstrated that aged WT animals were more susceptible to cisplatin-induced loss of mitochondrial proteins, including PGC1α, OPA1, cytochrome-C, and Cox IV. CONCLUSIONS: In our study, the negative effects of cisplatin were heighted in aged animals, whereas overexpression of PGC1α was sufficient to combat the neuromuscular dysfunction caused by cisplatin, especially in older animals. Hence, our observations indicate that aged animals may be more susceptible to develop chemotherapy side toxicities and that mitochondria-targeted strategies may serve as a tool to prevent chemotherapy-induced muscle wasting and weakness.


Assuntos
Antineoplásicos , Caquexia , Animais , Caquexia/etiologia , Cisplatino/efeitos adversos , Citocromos/metabolismo , Citocromos/farmacologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/farmacologia
20.
Aesthetic Plast Surg ; 46(5): 2517-2525, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35325306

RESUMO

BACKGROUND: Adipose browning occurs after white fat transfer. But its location and effects on fat graft survival remains controversial. This study was performed to locate the browning of fat grafts, and to explore the effects of quercetin on fat graft browning and fat graft survival. METHODS: Human fat granules were injected into the subcutaneous layer of 12 nude mice. Control group was injected with fat granules and 10% of normal saline, while quercetin group was injected with fat granules and 10% of quercetin. The graft samples (n = 6 for each group) were obtained in weeks 2, 4, 8 and 12. Weight retention rate of the grafts was calculated. Gene and protein expression of mitochondrial markers (silent information regulator 1, SIRT1; heat shock protein 60, HSP60), browning marker (uncoupling protein 1, UCP1), peroxisome proliferator-activated receptor-γ (PPAR-γ), vascular endothelial growth factor A (VEGF-A) were evaluated. Hematoxylin and eosin staining and anti-UCP1 staining were performed. RESULTS: Clusters of small multilocular beige adipocytes were observed in the periphery of fat grafts. Compared with control group, quercetin group had a higher weight retention rate, a higher gene/protein expression of SIRT1, HSP60, UCP1, PPAR-γ and VEGF-A, and a higher occurrence of peripheral adipose browning. CONCLUSIONS: Peripherally located adipose browning occurred after white fat transfer. It can be enhanced by the addition of quercetin through promoting mitochondrial function of fat cells, and may be one of the mechanisms that quercetin improves fat graft survival. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Assuntos
Sobrevivência de Enxerto , Fator A de Crescimento do Endotélio Vascular , Camundongos , Animais , Humanos , Proteína Desacopladora 1/genética , Quercetina/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/farmacologia , Camundongos Nus , Chaperonina 60/farmacologia , Sirtuína 1/farmacologia , Solução Salina/farmacologia , Hematoxilina/farmacologia , Amarelo de Eosina-(YS)/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA