Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Genes Dev ; 35(19-20): 1333-1338, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34531316

RESUMO

The full array of cold-responsive cell types within white adipose tissue that drive thermogenic beige adipocyte biogenesis remains undefined. We demonstrate that acute cold challenge elicits striking transcriptomic changes specifically within DPP4+ PDGFRß+ adipocyte precursor cells, including a ß-adrenergic receptor CREB-mediated induction in the expression of the prothermogenic cytokine, Il33 Doxycycline-inducible deletion of Il33 in PDGFRß+ cells at the onset of cold exposure attenuates ILC2 accumulation and beige adipocyte accrual. These studies highlight the multifaceted roles for adipocyte progenitors and the ability of select mesenchymal subpopulations to relay neuronal signals to tissue-resident immune cells in order to regulate tissue plasticity.


Assuntos
Adipócitos Bege , Adipócitos Bege/metabolismo , Tecido Adiposo Branco/metabolismo , Adrenérgicos/metabolismo , Temperatura Baixa , Imunidade Inata , Linfócitos , Termogênese/genética
2.
Genes Dev ; 35(21-22): 1461-1474, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34620682

RESUMO

Energy-storing white adipocytes maintain their identity by suppressing the energy-burning thermogenic gene program of brown and beige adipocytes. Here, we reveal that the protein-protein interaction between the transcriptional coregulator ZFP423 and brown fat determination factor EBF2 is essential for restraining the thermogenic phenotype of white adipose tissue (WAT). Disruption of the ZFP423-EBF2 protein interaction through CRISPR-Cas9 gene editing triggers widespread "browning" of WAT in adult mice. Mechanistically, ZFP423 recruits the NuRD corepressor complex to EBF2-bound thermogenic gene enhancers. Loss of adipocyte Zfp423 induces an EBF2 NuRD-to-BAF coregulator switch and a shift in PPARγ occupancy to thermogenic genes. This shift in PPARγ occupancy increases the antidiabetic efficacy of the PPARγ agonist rosiglitazone in obesity while diminishing the unwanted weight-gaining effect of the drug. These data indicate that ZFP423 controls EBF2 coactivator recruitment and PPARγ occupancy to determine the thermogenic plasticity of adipocytes and highlight the potential of therapeutically targeting transcriptional brakes to induce beige adipocyte biogenesis in obesity.


Assuntos
PPAR gama , Termogênese , Adipócitos Marrons/metabolismo , Adipócitos Brancos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA , Camundongos , PPAR gama/genética , Termogênese/genética , Fatores de Transcrição
3.
J Cell Physiol ; 236(2): 958-970, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32632982

RESUMO

Cellular senescence, a state of growth arrest, is involved in various age-related diseases. We previously found that carnitine palmitoyltransferase 1C (CPT1C) is a key regulator of cancer cell proliferation and senescence, but it is unclear whether CPT1C plays a similar role in normal cells. Therefore, this study aimed to investigate the role of CPT1C in cellular proliferation and senescence of human embryonic lung MRC-5 fibroblasts and the involved mechanisms. The results showed that CPT1C could reverse the cellular senescence of MRC-5 fibroblasts, as evidenced by reduced senescence-associated ß-galactosidase activity, downregulated messenger RNA (mRNA) expression of senescence-associated secretory phenotype factors, and enhanced bromodeoxyuridine incorporation. Lipidomics analysis further revealed that CPT1C gain-of-function reduced lipid accumulation and reversed abnormal metabolic reprogramming of lipids in late MRC-5 cells. Oil Red O staining and Nile red fluorescence also indicated significant reduction of lipid accumulation after CPT1C gain-of-function. Consequently, CPT1C gain-of-function significantly reversed mitochondrial dysfunction, as evaluated by increased adenosine triphosphate synthesis and mitochondrial transmembrane potential, decreased radical oxygen species, upregulated respiratory capacity and mRNA expression of genes related to mitochondrial function. In summary, CPT1C plays a vital role in MRC-5 cellular proliferation and can reverse MRC-5 cellular senescence through the regulation of lipid metabolism and mitochondrial function, which supports the role of CPT1C as a novel target for intervention into cellular proliferation and senescence and suggests CPT1C as a new strategy for antiaging.


Assuntos
Carnitina O-Palmitoiltransferase/genética , Senescência Celular/genética , Fibroblastos/fisiologia , Metabolismo dos Lipídeos/genética , Mitocôndrias/genética , Trifosfato de Adenosina/genética , Linhagem Celular , Proliferação de Células/genética , Regulação para Baixo/genética , Humanos , Potencial da Membrana Mitocondrial/genética , RNA Mensageiro/genética , Regulação para Cima/genética , beta-Galactosidase/genética
4.
Biochem Biophys Res Commun ; 515(4): 517-523, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31153640

RESUMO

Previous studies have suggested that nitric oxide (NO) which is synthetized by nitric oxide synthase (NOS) is closely related to the carcinogenesis and progression of colon cancer. However, the precise physiopathological role of NO on colon cancer remains unclear, and a lot of related studies focused on NOS2 and NOS3, but little on NOS1. Here, stable overexpression NOS1 of colon cancer cells were constructed to investigate whether NOS1 plays a special role in colon cancer. We observed that NOS1 protein was presented in mitochondria. Both the basal and cisplatin-induced mitochondrial superoxide were inhibited by NOS1, and the cisplatin-induced apoptosis was also inhibited by NOS1. Geldanamycin, a Hsp90 N-terminal inhibitor, was able to impede NOS1 translocation into mitochondria and reverse NOS1-induced apoptosis resistance. Importantly, SIRT3 activity was enhanced by NOS1, which contributes to the low level of mitochondrial superoxide and apoptosis resistance. Our data suggest a link between NOS1 and apoptosis resistance in colon cancer cells through mtNOS1-SIRT3-SOD2 axis. Furthermore, NOS1-induced apoptosis resistance could be reversed by inhibiting mitochondrial translocation of NOS1.


Assuntos
Apoptose , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Mitocôndrias/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Sirtuína 3/metabolismo , Benzoquinonas/farmacologia , Transporte Biológico , Caspase 3/metabolismo , Linhagem Celular Tumoral , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Lactamas Macrocíclicas/farmacologia , Nitritos/metabolismo , Isoformas de Proteínas , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
5.
ACS Omega ; 9(8): 8893-8903, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38434811

RESUMO

This experimental study investigates the impact of water injection into intake ports on combustion and emissions in gasoline engines. It also examines particle size distribution at various water-to-fuel ratios and explores the combined effects of water injection and compression ratios in gasoline engines. The results indicate that water injection effectively mitigates engine knock, reduces peak firing pressures, and moderates heat release rates through charge cooling. Advancing ignition timing with water injection advances combustion, resulting in reduced specific fuel consumption, particularly under moderate load conditions. Water injection lowers NOx emissions by reducing combustion temperatures but increases unburned THC emissions due to inhibited oxidation reaction rates. Minor effects were observed on CO emissions. Furthermore, particle numbers were significantly reduced with water injection, particularly in the nucleation mode particles. The simultaneous application of a higher compression ratio and water injection yields substantial improvements in fuel consumption with minimal impact on NOx and THC emissions.

6.
Cell Metab ; 35(3): 386-413, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36889280

RESUMO

Adipose tissue exhibits remarkable plasticity with capacity to change in size and cellular composition under physiological and pathophysiological conditions. The emergence of single-cell transcriptomics has rapidly transformed our understanding of the diverse array of cell types and cell states residing in adipose tissues and has provided insight into how transcriptional changes in individual cell types contribute to tissue plasticity. Here, we present a comprehensive overview of the cellular atlas of adipose tissues focusing on the biological insight gained from single-cell and single-nuclei transcriptomics of murine and human adipose tissues. We also offer our perspective on the exciting opportunities for mapping cellular transitions and crosstalk, which have been made possible by single-cell technologies.


Assuntos
Adipócitos , Adipogenia , Humanos , Animais , Camundongos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Perfilação da Expressão Gênica , Diferenciação Celular
7.
Cell Metab ; 34(5): 783-799.e7, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35447091

RESUMO

Single-cell RNA sequencing (scRNA-seq) has revealed that adult white adipose tissue (WAT) harbors functionally diverse subpopulations of mesenchymal stromal cells that differentially impact tissue plasticity. To date, the molecular basis of this cellular heterogeneity has not been fully defined. Here, we describe a multilayered omics approach to dissect adipose progenitor cell heterogeneity in three dimensions: progenitor subpopulation, sex, and anatomical localization. We applied state-of-the-art mass spectrometry methods to quantify 4,870 proteins in eight different stromal cell populations from perigonadal and inguinal WAT of male and female mice and acquired transcript expression levels of 15,477 genes using RNA-seq. Our data unveil molecular signatures defining sex differences in preadipocyte differentiation and identify regulatory pathways that functionally distinguish adipose progenitor subpopulations. This multilayered omics analysis, freely accessible at http://preadprofiler.net/, provides unprecedented insights into adipose stromal cell heterogeneity and highlights the benefit of complementary proteomics to support findings from scRNA-seq studies.


Assuntos
Adipócitos , Adipogenia , Adipócitos/metabolismo , Tecido Adiposo , Tecido Adiposo Branco/metabolismo , Animais , Diferenciação Celular , Feminino , Masculino , Camundongos , Células-Tronco/metabolismo
8.
Nat Metab ; 4(8): 1055-1070, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35982290

RESUMO

Adult white adipose tissue (WAT) harbors distinct mesenchymal stromal cell subpopulations that differentially affect WAT function and plasticity. Here we unveil the cellular landscape of the perinatal epididymal WAT primordium using single-cell transcriptomics in male mice. We reveal that adipocyte precursor cells and fibro-inflammatory progenitors (FIPs) emerge as functionally distinct PDGFRß+ subpopulations within the epididymal WAT anlagen prior to adipocyte accrual. We further identify important molecular and functional differences between perinatal and adult FIPs, including differences in their pro-inflammatory response, adipogenic capacity and anti-adipogenic behavior. Notably, we find that transient overexpression of Pparg in PDGFRß+ cells only during postnatal days 0.5 to 7.5 in male mice leads to hyperplastic WAT development, durable progenitor cell reprogramming, and protection against pathologic WAT remodeling and glucose intolerance in adult-onset obesity. Thus, factors that alter the adipogenic capacity of perinatal adipose progenitors can have long-lasting effects on progenitor plasticity, tissue expandability and metabolic health into adulthood.


Assuntos
Adipogenia , Tecido Adiposo Branco , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Feminino , Masculino , Camundongos , Obesidade/metabolismo , Gravidez , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
9.
J Clin Invest ; 132(21)2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36066975

RESUMO

The molecular mechanisms underlying obesity-induced increases in ß cell mass and the resulting ß cell dysfunction need to be elucidated further. Our study revealed that GPR92, expressed in islet macrophages, is modulated by dietary interventions in metabolic tissues. Therefore, we aimed to define the role of GPR92 in islet inflammation by using a high-fat diet-induced (HFD-induced) obese mouse model. GPR92-KO mice exhibited glucose intolerance and reduced insulin levels - despite the enlarged pancreatic islets - as well as increased islet macrophage content and inflammation level compared with WT mice. These results indicate that the lack of GPR92 in islet macrophages can cause ß cell dysfunction, leading to disrupted glucose homeostasis. Alternatively, stimulation with the GPR92 agonist farnesyl pyrophosphate results in the inhibition of HFD-induced islet inflammation and increased insulin secretion in WT mice, but not in GPR92-KO mice. Thus, our study suggests that GPR92 can be a potential target to alleviate ß cell dysfunction via the inhibition of islet inflammation associated with the progression of diabetes.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Camundongos , Animais , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Obesidade/metabolismo , Ilhotas Pancreáticas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Camundongos Obesos , Macrófagos/metabolismo , Inflamação/metabolismo , Camundongos Endogâmicos C57BL
10.
Cell Rep ; 40(11): 111362, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36103820

RESUMO

Obesity is associated with increased cancer incidence and progression. However, the relationship between adiposity and cancer remains poorly understood at the mechanistic level. Here, we report that adipocytes from tumor-invasive mammary fat undergo de-differentiation to fibroblast-like precursor cells during tumor progression and integrate into the tumor microenvironment. Single-cell sequencing reveals that these de-differentiated adipocytes lose their original identities and transform into multiple cell types, including myofibroblast- and macrophage-like cells, with their characteristic features involved in immune response, inflammation, and extracellular matrix remodeling. The de-differentiated cells are metabolically distinct from tumor-associated fibroblasts but exhibit comparable effects on tumor cell proliferation. Inducing de-differentiation by Xbp1s overexpression promotes tumor progression despite lower adiposity. In contrast, promoting lipid-storage capacity in adipocytes through MitoNEET overexpression curbs tumor growth despite greater adiposity. Collectively, the metabolic interplay between tumor cells and adipocytes induces adipocyte mesenchymal transition and contributes to reconfigure the stroma into a more tumor-friendly microenvironment.


Assuntos
Neoplasias da Mama , Neoplasias Mamárias Animais , Adipócitos/metabolismo , Animais , Neoplasias da Mama/patologia , Matriz Extracelular/metabolismo , Feminino , Humanos , Neoplasias Mamárias Animais/patologia , Microambiente Tumoral
11.
Cell Stem Cell ; 28(4): 702-717.e8, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33539722

RESUMO

The adipose tissue stroma is a rich source of molecularly distinct stem and progenitor cell populations with diverse functions in metabolic regulation, adipogenesis, and inflammation. The ontology of these populations and the mechanisms that govern their behaviors in response to stimuli, such as overfeeding, however, are unclear. Here, we show that the developmental fates and functional properties of adipose platelet-derived growth factor receptor beta (PDGFRß)+ progenitor subpopulations are tightly regulated by mitochondrial metabolism. Reducing the mitochondrial ß-oxidative capacity of PDGFRß+ cells via inducible expression of MitoNEET drives a pro-inflammatory phenotype in adipose progenitors and alters lineage commitment. Furthermore, disrupting mitochondrial function in PDGFRß+ cells rapidly induces alterations in immune cell composition in lean mice and impacts expansion of adipose tissue in diet-induced obesity. The adverse effects on adipose tissue remodeling can be reversed by restoring mitochondrial activity in progenitors, suggesting therapeutic potential for targeting energy metabolism in these cells.


Assuntos
Adipogenia , Tecido Adiposo Branco , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Proteínas de Ligação ao Ferro/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias , Células-Tronco/metabolismo
12.
Cell Stem Cell ; 28(4): 685-701.e7, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33539723

RESUMO

Adipose precursor cells (APCs) exhibit regional variation in response to obesity, for unclear reasons. Here, we reveal that HIFα-induced PDGFRß signaling within murine white adipose tissue (WAT) PDGFRß+ cells drives inhibitory serine 112 (S112) phosphorylation of PPARγ, the master regulator of adipogenesis. Levels of PPARγ S112 phosphorylation in WAT PDGFRß+ cells are depot dependent, with levels of PPARγ phosphorylation in PDGFRß+ cells inversely correlating with their capacity for adipogenesis upon high-fat-diet feeding. HIFα suppression in PDGFRß+ progenitors promotes subcutaneous and intra-abdominal adipogenesis, healthy WAT remodeling, and improved metabolic health in obesity. These metabolic benefits are mimicked by treatment of obese mice with the PDGFR antagonist Imatinib, which promotes adipocyte hyperplasia and glucose tolerance in a progenitor cell PPARγ-dependent manner. Our studies unveil a mechanism underlying depot-specific responses of APCs to high-fat feeding and highlight the potential for APCs to be targeted pharmacologically to improve metabolic health in obesity.


Assuntos
Adipogenia , Tecido Adiposo , Adipócitos , Tecido Adiposo Branco , Animais , Dieta Hiperlipídica , Camundongos , Camundongos Endogâmicos C57BL , Obesidade
13.
J Vis Exp ; (162)2020 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-32865529

RESUMO

The stromal-vascular fraction (SVF) of white adipose tissue (WAT) is remarkably heterogeneous and consists of numerous cell types that contribute functionally to the expansion and remodeling of WAT in adulthood. A tremendous barrier to studying the implications of this cellular heterogeneity is the inability to readily isolate functionally distinct cell subpopulations from WAT SVF for in vitro and in vivo analyses. Single-cell sequencing technology has recently identified functionally distinct fibro-inflammatory and adipogenic PDGFRß+ perivascular cell subpopulations in intra-abdominal WAT depots of adult mice. Fibro-inflammatory progenitors (termed, "FIPs") are non-adipogenic collagen producing cells that can exert a pro-inflammatory phenotype. PDGFRß+ adipocyte precursor cells (APCs) are highly adipogenic both in vitro and in vivo upon cell transplantation. Here, we describe multiple methods for the isolation of these stromal cell subpopulations from murine intra-abdominal WAT depots. FIPs and APCs can be isolated by fluorescence-activated cell sorting (FACS) or by taking advantage of biotinylated antibody-based immunomagnetic bead technology. Isolated cells can be used for molecular and functional analysis. Studying the functional properties of stromal cell subpopulation in isolation will expand our current knowledge of adipose tissue remodeling under physiological or pathological conditions on the cellular level.


Assuntos
Gordura Abdominal/citologia , Adipogenia , Separação Celular/métodos , Células Estromais/citologia , Tecido Adiposo Branco/citologia , Animais , Citometria de Fluxo , Inflamação/patologia , Camundongos , Células Estromais/patologia
14.
Nat Metab ; 2(11): 1332-1349, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33139957

RESUMO

Chronic low-grade white adipose tissue (WAT) inflammation is a hallmark of metabolic syndrome in obesity. Here, we demonstrate that a subpopulation of mouse WAT perivascular (PDGFRß+) cells, termed fibro-inflammatory progenitors (FIPs), activate proinflammatory signalling cascades shortly after the onset of high-fat diet feeding and regulate proinflammatory macrophage accumulation in WAT in a TLR4-dependent manner. FIPs activation in obesity is mediated by the downregulation of zinc-finger protein 423 (ZFP423), identified here as a transcriptional corepressor of NF-κB. ZFP423 suppresses the DNA-binding capacity of the p65 subunit of NF-κB by inducing a p300-to-NuRD coregulator switch. Doxycycline-inducible expression of Zfp423 in PDGFRß+ cells suppresses inflammatory signalling in FIPs and attenuates metabolic inflammation of visceral WAT in obesity. Inducible inactivation of Zfp423 in PDGFRß+ cells increases FIP activity, exacerbates adipose macrophage accrual and promotes WAT dysfunction. These studies implicate perivascular mesenchymal cells as important regulators of chronic adipose-tissue inflammation in obesity and identify ZFP423 as a transcriptional break on NF-κB signalling.


Assuntos
Tecido Adiposo Branco/patologia , Macrófagos/patologia , Células-Tronco Mesenquimais , Obesidade/patologia , Animais , Proteínas de Ligação a DNA/metabolismo , Dieta Hiperlipídica , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/metabolismo
15.
Elife ; 72018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30265241

RESUMO

White adipose tissue (WAT) remodeling is dictated by coordinated interactions between adipocytes and resident stromal-vascular cells; however, the functional heterogeneity of adipose stromal cells has remained unresolved. We combined single-cell RNA-sequencing and FACS to identify and isolate functionally distinct subpopulations of PDGFRß+ stromal cells within visceral WAT of adult mice. LY6C- CD9- PDGFRß+ cells represent highly adipogenic visceral adipocyte precursor cells ('APCs'), whereas LY6C+ PDGFRß+ cells represent fibro-inflammatory progenitors ('FIPs'). FIPs lack adipogenic capacity, display pro-fibrogenic/pro-inflammatory phenotypes, and can exert an anti-adipogenic effect on APCs. The pro-inflammatory phenotype of PDGFRß+ cells is regulated, at least in part, by NR4A nuclear receptors. These data highlight the functional heterogeneity of visceral WAT perivascular cells, and provide insight into potential cell-cell interactions impacting adipogenesis and inflammation. These improved strategies to isolate FIPs and APCs from visceral WAT will facilitate the study of physiological WAT remodeling and mechanisms leading to metabolic dysfunction. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed.


Assuntos
Adipogenia , Envelhecimento/patologia , Inflamação/patologia , Gordura Intra-Abdominal/patologia , Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Antígenos Ly/metabolismo , Diferenciação Celular , Separação Celular , Dieta Hiperlipídica , Feminino , Fibrose , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fenótipo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Células Estromais/metabolismo , Células Estromais/patologia , Tetraspanina 29/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA