Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Metab ; 35(12): 2107-2118.e6, 2023 12 05.
Article in English | MEDLINE | ID: mdl-37863051

ABSTRACT

Fructose consumption is associated with tumor growth and metastasis in mice, yet its impact on antitumor immune responses remains unclear. Here, we show that dietary fructose modulates adipocyte metabolism to enhance antitumor CD8+ T cell immune responses and control tumor growth. Transcriptional profiling of tumor-infiltrating CD8+ T cells reveals that dietary fructose mediates attenuated transition of CD8+ T cells to terminal exhaustion, leading to a superior antitumor efficacy. High-fructose feeding initiates adipocyte-derived leptin production in an mTORC1-dependent manner, thereby triggering leptin-boosted antitumor CD8+ T cell responses. Importantly, high plasma leptin levels are correlated with elevated plasma fructose concentrations and improved antitumor CD8+ T cell responses in patients with lung cancer. Our study characterizes a critical role for dietary fructose in shaping adipocyte metabolism to prime antitumor CD8+ T cell responses and highlights that the fructose-leptin axis may be harnessed for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Mice , Animals , Leptin/metabolism , Neoplasms/metabolism , Immunotherapy , Lymphocyte Activation
2.
Redox Rep ; 28(1): 2260646, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37750478

ABSTRACT

Primary hepatocytes are widely used as a tool for studying metabolic function and regulation in the liver. However, the metabolic properties of primary hepatocytes are gradually lost after isolation. Here, we illustrated that fatty acid metabolism is the major compromised metabolic process in isolated primary hepatocytes, along with drastically decreased GSH and ROS content, while lipid peroxidation is increased. Gain- and loss-of-function studies revealed that Slc7a11 expression is critical in maintaining fatty acid metabolism and facilitating hormone-induced fatty acid metabolic events, which is synergistic with dexamethasone treatment. Intriguingly, Slc7a11 expression and dexamethasone treatment cooperatively upregulated AKT and AMPK signaling and mitochondrial complex expression in primary hepatocytes. Furthermore, direct treatment with reduced GSH or inhibition of ferroptosis is sufficient to drive protective effects on fatty acid metabolism in primary hepatocytes. Our results demonstrate that Slc7a11 expression in isolated primary hepatocytes induces GSH production, which protects against ferroptosis, to increase fatty acid metabolic gene expression, AKT and AMPK signaling and mitochondrial function in synergy with dexamethasone treatment, thereby efficiently preserving primary hepatocyte metabolic signatures, thus providing a promising approach to better reserve primary hepatocyte metabolic activities after isolation to potentially improve the understanding of liver biological functions from studies using primary hepatocytes.


Subject(s)
AMP-Activated Protein Kinases , Proto-Oncogene Proteins c-akt , Hepatocytes , Fatty Acids , Dexamethasone/pharmacology , Glutathione
3.
Adv Sci (Weinh) ; 10(25): e2207549, 2023 09.
Article in English | MEDLINE | ID: mdl-37401236

ABSTRACT

LncRNAs play a critical role in oral squamous cell carcinoma (OSCC) progression. However, the function and detailed molecular mechanism of most lncRNAs in OSCC are not fully understood. Here, a novel nuclear-localized lncRNA, DUXAP9 (DUXAP9), that is highly expressed in OSCC is identified. A high level of DUXAP9 is positively associated with lymph node metastasis, poor pathological differentiation, advanced clinical stage, worse overall survival, and worse disease-specific survival in OSCC patients. Overexpression of DUXAP9 significantly promotes OSCC cell proliferation, migration, invasion, and xenograft tumor growth and metastasis, and upregulates N-cadherin, Vimentin, Ki67, PCNA, and EZH2 expression and downregulates E-cadherin in vitro and in vivo, whereas knockdown of DUXAP9 remarkably suppresses OSCC cell proliferation, migration, invasion, and xenograft tumor growth in vitro and in vivo in an EZH2-dependent manner. Yin Yang 1 (YY1) is found to activate the transcriptional expression of DUXAP9 in OSCC. Furthermore, DUXAP9 physically interacts with EZH2 and inhibits EZH2 degradation via the suppression of EZH2 phosphorylation, thereby blocking EZH2 translocation from the nucleus to the cytoplasm. Thus, DUXAP9 can serve as a promising target for OSCC therapy.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Squamous Cell/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Yin-Yang , Cell Line, Tumor , Cell Proliferation/genetics , Mouth Neoplasms/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , CDC2 Protein Kinase
4.
Nat Commun ; 11(1): 2379, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32404872

ABSTRACT

Brown and beige fat share a remarkably similar transcriptional program that supports fuel oxidation and thermogenesis. The chromatin-remodeling machinery that governs genome accessibility and renders adipocytes poised for thermogenic activation remains elusive. Here we show that BAF60a, a subunit of the SWI/SNF chromatin-remodeling complexes, serves an indispensable role in cold-induced thermogenesis in brown fat. BAF60a maintains chromatin accessibility at PPARγ and EBF2 binding sites for key thermogenic genes. Surprisingly, fat-specific BAF60a inactivation triggers more pronounced cold-induced browning of inguinal white adipose tissue that is linked to induction of MC2R, a receptor for the pituitary hormone ACTH. Elevated MC2R expression sensitizes adipocytes and BAF60a-deficient adipose tissue to thermogenic activation in response to ACTH stimulation. These observations reveal an unexpected dichotomous role of BAF60a-mediated chromatin remodeling in transcriptional control of brown and beige gene programs and illustrate a pituitary-adipose signaling axis in the control of thermogenesis.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Cold Temperature , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, Brown/ultrastructure , Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Adrenocorticotropic Hormone/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , Cells, Cultured , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Expression/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Thermogenesis/drug effects , Thermogenesis/genetics
5.
Sci Adv ; 6(20): eaay6191, 2020 05.
Article in English | MEDLINE | ID: mdl-32426492

ABSTRACT

Depletion of fat-resident regulatory T cells (Tregs) and group 2 innate lymphoid cells (ILC2s) has been causally linked to obesity-associated insulin resistance. However, the molecular nature of the pathogenic signals suppress adipose Tregs and ILC2s in obesity remains unknown. Here, we identified the soluble isoform of interleukin (IL)-33 receptor ST2 (sST2) as an obesity-induced adipokine that attenuates IL-33 signaling and disrupts Treg/ILC2 homeostasis in adipose tissue, thereby exacerbates obesity-associated insulin resistance in mice. We demonstrated sST2 is a target of TNFα signaling in adipocytes that is countered by Zbtb7b. Fat-specific ablation of Zbtb7b augments adipose sST2 gene expression, leading to diminished fat-resident Tregs/ILC2s, more pronounced adipose tissue inflammation and fibrosis, and impaired glucose homeostasis in mice. Mechanistically, Zbtb7b suppresses NF-κB activation in response to TNFα through destabilizing IκBα. These findings uncover an adipokine-immune signaling pathway that is engaged in obesity to drive the pathological changes of the immunometabolic landscape.


Subject(s)
Insulin Resistance , Adipokines/metabolism , Adipose Tissue/metabolism , Animals , DNA-Binding Proteins/metabolism , Immunity, Innate , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Cell ; 75(3): 644-660.e5, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398325

ABSTRACT

Cell-cell communication via ligand-receptor signaling is a fundamental feature of complex organs. Despite this, the global landscape of intercellular signaling in mammalian liver has not been elucidated. Here we perform single-cell RNA sequencing on non-parenchymal cells isolated from healthy and NASH mouse livers. Secretome gene analysis revealed a highly connected network of intrahepatic signaling and disruption of vascular signaling in NASH. We uncovered the emergence of NASH-associated macrophages (NAMs), which are marked by high expression of triggering receptors expressed on myeloid cells 2 (Trem2), as a feature of mouse and human NASH that is linked to disease severity and highly responsive to pharmacological and dietary interventions. Finally, hepatic stellate cells (HSCs) serve as a hub of intrahepatic signaling via HSC-derived stellakines and their responsiveness to vasoactive hormones. These results provide unprecedented insights into the landscape of intercellular crosstalk and reprogramming of liver cells in health and disease.


Subject(s)
Cell Communication/genetics , Liver/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Sequence Analysis, RNA , Animals , Cellular Reprogramming/genetics , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Ligands , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/genetics , Single-Cell Analysis
7.
Nat Commun ; 9(1): 2986, 2018 07 30.
Article in English | MEDLINE | ID: mdl-30061575

ABSTRACT

Hepatic lipogenesis is aberrantly induced in nonalcoholic fatty liver disease (NAFLD) via activation of the LXR-SREBP1c pathway. To date, a number of protein factors impinging on the transcriptional activity of LXR and SREBP1c have been elucidated. However, whether this regulatory axis interfaces with long noncoding RNAs (lncRNAs) remains largely unexplored. Here we show that hepatic expression of the lncRNA Blnc1 is strongly elevated in obesity and NAFLD in mice. Blnc1 is required for the induction of SREBP1c and hepatic lipogenic genes in response to LXR activation. Liver-specific inactivation of Blnc1 abrogates high-fat diet-induced hepatic steatosis and insulin resistance and protects mice from diet-induced nonalcoholic steatohepatitis. Proteomic analysis of the Blnc1 ribonucleoprotein complex identified EDF1 as a component of the LXR transcriptional complex that acts in concert with Blnc1 to activate the lipogenic gene program. These findings illustrate a lncRNA transcriptional checkpoint that licenses excess hepatic lipogenesis to exacerbate insulin resistance and NAFLD.


Subject(s)
Lipogenesis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Obesity/complications , RNA, Long Noncoding/genetics , Adipose Tissue/metabolism , Animals , Bile Acids and Salts/chemistry , CRISPR-Cas Systems , Calmodulin-Binding Proteins/metabolism , Disease Models, Animal , Fatty Liver , Gene Expression Profiling , HEK293 Cells , Hepatocytes/metabolism , Humans , Insulin Resistance , Liver/physiopathology , Liver X Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/genetics , Protein Interaction Mapping , Proteomics , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription, Genetic
8.
Mol Metab ; 14: 60-70, 2018 08.
Article in English | MEDLINE | ID: mdl-29934059

ABSTRACT

OBJECTIVE: Long noncoding RNAs (lncRNAs) are emerging as powerful regulators of adipocyte differentiation and gene expression. However, their significance in adipose tissue metabolism and physiology has not been demonstrated in vivo. We previously identified Blnc1 as a conserved lncRNA regulator of brown and beige adipocyte differentiation. In this study, we investigated the physiological role of Blnc1 in thermogenesis, adipose remodeling and systemic metabolism. METHODS: We generated fat-specific Blnc1 transgenic and conditional knockout mouse strains and investigated how adipocyte Blnc1 levels are causally linked to key aspects of metabolic health following diet-induced obesity. We performed studies using cultured adipocytes to establish cell-autonomous role of Blnc1 in regulating adipocyte gene programs. RESULTS: Blnc1 is highly induced in both brown and white fats from obese mice. Fat-specific inactivation of Blnc1 impairs cold-induced thermogenesis and browning and exacerbates obesity-associated brown fat whitening, adipose tissue inflammation and fibrosis, leading to more severe insulin resistance and hepatic steatosis. On the contrary, transgenic expression of Blnc1 in adipose tissue elicits the opposite and beneficial metabolic effects, supporting a critical role of Blnc1 in driving adipose adaptation and homeostatic remodeling during obesity. Mechanistically, Blnc1 cell-autonomously attenuates proinflammatory cytokine signaling and promotes fuel storage in adipocytes through its protein partner Zbtb7b. CONCLUSIONS: This study illustrates a surprisingly pleiotropic and dominant role of lncRNA in driving adaptive adipose tissue remodeling and preserving metabolic health.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Homeostasis , Obesity/genetics , RNA, Long Noncoding/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Proc Natl Acad Sci U S A ; 114(34): E7111-E7120, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28784777

ABSTRACT

Brown and beige adipocytes convert chemical energy into heat through uncoupled respiration to defend against cold stress. Beyond thermogenesis, brown and beige fats engage other metabolic tissues via secreted factors to influence systemic energy metabolism. How the protein and long noncoding RNA (lncRNA) regulatory networks act in concert to regulate key aspects of thermogenic adipocyte biology remains largely unknown. Here we developed a genome-wide functional screen to interrogate the transcription factors and cofactors in thermogenic gene activation and identified zinc finger and BTB domain-containing 7b (Zbtb7b) as a potent driver of brown fat development and thermogenesis and cold-induced beige fat formation. Zbtb7b is required for activation of the thermogenic gene program in brown and beige adipocytes. Genetic ablation of Zbtb7b impaired cold-induced transcriptional remodeling in brown fat, rendering mice sensitive to cold temperature, and diminished browning of inguinal white fat. Proteomic analysis revealed a mechanistic link between Zbtb7b and the lncRNA regulatory pathway through which Zbtb7b recruits the brown fat lncRNA 1 (Blnc1)/heterogeneous nuclear ribonucleoprotein U (hnRNPU) ribonucleoprotein complex to activate thermogenic gene expression in adipocytes. These findings illustrate the emerging concept of a protein-lncRNA regulatory network in the control of adipose tissue biology and energy metabolism.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, Brown/metabolism , DNA-Binding Proteins/metabolism , Thermogenesis , Transcription Factors/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Beige/growth & development , Adipose Tissue, Brown/growth & development , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Long Noncoding , Transcription Factors/genetics
10.
Mol Metab ; 6(8): 863-872, 2017 08.
Article in English | MEDLINE | ID: mdl-28752050

ABSTRACT

OBJECTIVE: Brown and white adipose tissue exerts pleiotropic effects on systemic energy metabolism in part by releasing endocrine factors. Neuregulin 4 (Nrg4) was recently identified as a brown fat-enriched secreted factor that ameliorates diet-induced metabolic disorders, including insulin resistance and hepatic steatosis. However, the physiological mechanisms through which Nrg4 regulates energy balance and glucose and lipid metabolism remain incompletely understood. The aims of the current study were: i) to investigate the regulation of adipose Nrg4 expression during obesity and the physiological signals involved, ii) to elucidate the mechanisms underlying Nrg4 regulation of energy balance and glucose and lipid metabolism, and iii) to explore whether Nrg4 regulates adipose tissue secretome gene expression and adipokine secretion. METHODS: We examined the correlation of adipose Nrg4 expression with obesity in a cohort of diet-induced obese mice and investigated the upstream signals that regulate Nrg4 expression. We performed metabolic cage and hyperinsulinemic-euglycemic clamp studies in Nrg4 transgenic mice to dissect the metabolic pathways regulated by Nrg4. We investigated how Nrg4 regulates hepatic lipid metabolism in the fasting state and explored the effects of Nrg4 on adipose tissue gene expression, particularly those encoding secreted factors. RESULTS: Adipose Nrg4 expression is inversely correlated with adiposity and regulated by pro-inflammatory and anti-inflammatory signaling. Transgenic expression of Nrg4 increases energy expenditure and augments whole body glucose metabolism. Nrg4 protects mice from diet-induced hepatic steatosis in part through activation of hepatic fatty acid oxidation and ketogenesis. Finally, Nrg4 promotes a healthy adipokine profile during obesity. CONCLUSIONS: Nrg4 exerts pleiotropic beneficial effects on energy balance and glucose and lipid metabolism to ameliorate obesity-associated metabolic disorders. Biologic therapeutics based on Nrg4 may improve both type 2 diabetes and non-alcoholic fatty liver disease (NAFLD) in patients.


Subject(s)
Adipokines/blood , Fatty Acids/metabolism , Neuregulins/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Adipocytes/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Energy Metabolism , Glucose/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Neuregulins/genetics , Non-alcoholic Fatty Liver Disease/etiology , Obesity/etiology
11.
Mol Metab ; 6(1): 101-110, 2017 01.
Article in English | MEDLINE | ID: mdl-28123941

ABSTRACT

OBJECTIVE: Long noncoding RNAs (lncRNAs) are emerging as important regulators of diverse biological processes. Recent work has demonstrated that the inducible lncRNA Blnc1 stimulates thermogenic gene expression during brown and beige adipocyte differentiation. However, whether Blnc1 is functionally conserved in humans has not been explored. In addition, the molecular basis of the Blnc1 ribonucleoprotein complex in thermogenic gene induction remains incompletely understood. The aims of the current study were to: i) investigate functional conservation of Blnc1 in mice and humans and ii) elucidate the molecular mechanisms by which Blnc1 controls the thermogenic gene program in brown adipocytes. METHODS: Full-length human Blnc1 was cloned and examined for its ability to stimulate brown adipocyte differentiation. Different truncation mutants of Blnc1 were generated to identify functional RNA domains responsible for thermogenic gene induction. RNA-protein interaction studies were performed to delineate the molecular features of the Blnc1 ribonucleoprotein complex. RESULTS: Blnc1 is highly conserved in mice and humans at the sequence and function levels, both capable of stimulating brown adipocyte gene expression. A conserved RNA domain was identified to be required and sufficient for the biological activity of Blnc1. We identified hnRNPU as an RNA-binding protein that facilitates the assembly and augments the transcriptional function of the Blnc1/EBF2 ribonucleoprotein complex. CONCLUSIONS: Blnc1 is a conserved lncRNA that promotes thermogenic gene expression in brown adipocytes through formation of the Blnc1/hnRNPU/EBF2 ribonucleoprotein complex.


Subject(s)
Adipocytes, Brown/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation/physiology , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , Mice , RNA Recognition Motif Proteins , RNA, Long Noncoding/metabolism , Ribonucleoproteins/physiology , Thermogenesis/genetics , Transcription Factors/genetics
12.
EMBO Rep ; 16(11): 1563-80, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26415504

ABSTRACT

Apoptosis-inducing factor (AIF) exerts dual roles on cell death and survival, but its substrates as a putative oxidoreductase and roles in tumorigenesis remain elusive. Here, we report that AIF physically interacts with and inhibits the oxidation of phosphatase and tensin homolog on chromosome ten (PTEN), a tumor suppressor susceptible for oxidation-mediated inactivation. More intriguingly, we also identify PTEN as a mitochondrial protein and the ectopic expression of mitochondrial targeting sequence-carrying PTEN almost completely inhibits Akt phosphorylation in PTEN-deficient cells. AIF knockdown causes oxidation-mediated inactivation of the lipid phosphatase activity of PTEN, with ensuing activation of Akt kinase, phosphorylation of the Akt substrate GSK-3ß, and activation of ß-catenin signaling in cancer cells. Through its effect on ß-catenin signaling, AIF inhibits epithelial-mesenchymal transition (EMT) and metastasis of cancer cells in vitro and in orthotopically implanted xenografts. Accordingly, the expression of AIF is correlated with the survival of human patients with cancers of multiple origins. These results identify PTEN as the substrate of AIF oxidoreductase and reveal a novel function for AIF in controlling tumor metastasis.


Subject(s)
Apoptosis Inducing Factor/metabolism , Neoplasm Metastasis/physiopathology , PTEN Phosphohydrolase/metabolism , Protein Interaction Domains and Motifs , beta Catenin/metabolism , Apoptosis Inducing Factor/genetics , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Heterografts , Humans , Mitochondria/chemistry , Oxidation-Reduction , Oxidoreductases/metabolism , PTEN Phosphohydrolase/genetics , Phosphorylation
13.
Trends Biochem Sci ; 40(10): 586-596, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26410599

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as an integral part of the regulatory information encoded in the genome. lncRNAs possess the unique capability to interact with nucleic acids and proteins, and exert discrete effects on numerous biological processes. Recent studies have delineated multiple lncRNA pathways that control metabolic tissue development and function. The expansion of the regulatory code that links nutrient and hormonal signals to tissue metabolism gives new insights into the genetic and pathogenic mechanisms underlying metabolic disease. This review discusses lncRNA biology with a focus on their role in the development, signaling, and function of key metabolic tissues.


Subject(s)
RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Humans
14.
Trends Endocrinol Metab ; 26(5): 231-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25843910

ABSTRACT

Brown fat is highly active in fuel oxidation and dissipates chemical energy through uncoupling protein (UCP)1-mediated heat production. Activation of brown fat leads to increased energy expenditure, reduced adiposity, and lower plasma glucose and lipid levels, thus contributing to better homeostasis. Uncoupled respiration and thermogenesis have been considered to be responsible for the metabolic benefits of brown adipose tissue. Recent studies have demonstrated that brown adipocytes also secrete factors that act locally and systemically to influence fuel and energy metabolism. This review discusses the evidence supporting a thermogenesis-independent role of brown fat, particularly through its release of secreted factors, and their implications in physiology and therapeutic development.


Subject(s)
Adipose Tissue, Brown/metabolism , Thermogenesis , Adipocytes, Brown/metabolism , Adiponectin/physiology , Adipose Tissue, Brown/innervation , Animals , Bone Morphogenetic Proteins/physiology , Energy Metabolism , Fibroblast Growth Factors/physiology , Homeostasis , Humans , Ion Channels/physiology , Mitochondrial Proteins/physiology , Nerve Growth Factor/physiology , Neuregulins/physiology , Obesity , Uncoupling Protein 1 , Vascular Endothelial Growth Factor A/physiology
15.
Nat Med ; 20(12): 1436-1443, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401691

ABSTRACT

Brown fat activates uncoupled respiration in response to cold temperature and contributes to systemic metabolic homeostasis. To date, the metabolic action of brown fat has been primarily attributed to its role in fuel oxidation and uncoupling protein 1 (UCP1)-mediated thermogenesis. Whether brown fat engages other tissues through secreted factors remains largely unexplored. Here we show that neuregulin 4 (Nrg4), a member of the epidermal growth factor (EGF) family of extracellular ligands, is highly expressed in adipose tissues, enriched in brown fat and markedly increased during brown adipocyte differentiation. Adipose tissue Nrg4 expression was reduced in rodent and human obesity. Gain- and loss-of-function studies in mice demonstrated that Nrg4 protects against diet-induced insulin resistance and hepatic steatosis through attenuating hepatic lipogenic signaling. Mechanistically, Nrg4 activates ErbB3 and ErbB4 signaling in hepatocytes and negatively regulates de novo lipogenesis mediated by LXR and SREBP1c in a cell-autonomous manner. These results establish Nrg4 as a brown fat-enriched endocrine factor with therapeutic potential for the treatment of obesity-associated disorders, including type 2 diabetes and nonalcoholic fatty liver disease (NAFLD).


Subject(s)
Adipose Tissue, Brown/metabolism , Lipogenesis , Liver/metabolism , Neuregulins/genetics , Obesity/genetics , RNA, Messenger/genetics , Receptor, ErbB-3/metabolism , Receptor, ErbB-4/metabolism , 3T3-L1 Cells , Adipocytes, Brown/metabolism , Adipogenesis , Animals , Fatty Liver/genetics , Fatty Liver/metabolism , HEK293 Cells , Humans , Insulin Resistance , Liver X Receptors , Mice , Neuregulins/metabolism , Obesity/metabolism , Orphan Nuclear Receptors/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
16.
Mol Cell ; 55(3): 372-82, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25002143

ABSTRACT

Brown and beige/brite fats generate heat via uncoupled respiration to defend against cold. The total mass and activity of thermogenic adipose tissues are also tightly linked to systemic energy and nutrient homeostasis. Despite originating from distinct progenitors, brown and beige adipocytes acquire remarkably similar molecular and metabolic characteristics during differentiation through the action of a network of transcription factors and cofactors. How this regulatory network interfaces with long noncoding RNAs (lncRNAs), an emerging class of developmental regulators, remains largely unexplored. Here, we globally profiled lncRNA gene expression during thermogenic adipocyte formation and identified Brown fat lncRNA 1 (Blnc1) as a nuclear lncRNA that promotes brown and beige adipocyte differentiation and function. Blnc1 forms a ribonucleoprotein complex with transcription factor EBF2 to stimulate the thermogenic gene program. Further, Blnc1 itself is a target of EBF2, thereby forming a feedforward regulatory loop to drive adipogenesis toward thermogenic phenotype.


Subject(s)
Adipogenesis , Adipose Tissue, Brown/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , RNA, Long Noncoding/metabolism , Animals , Cell Line , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , Ribonucleoproteins/metabolism
17.
Int J Biochem Mol Biol ; 3(2): 165-78, 2012.
Article in English | MEDLINE | ID: mdl-22773957

ABSTRACT

Heterodimeric transcription factor hypoxia inducible factor-1 (HIF-1) functions as a master regulator of oxygen homeostasis in almost all nucleated mammalian cells. The fundamental process adapted to cellular oxygen alteration largely depends on the refined regulation on its alpha subunit, HIF-1α. Recent studies have unraveled expanding and critical roles of HIF-1α, involving in a multitude of developmental, physiological, and pathophysiological processes. This review will focus on the current knowledge of HIF-1α-targeting genes and its interacting proteins, as well as the concomitant functional relationships between them.

18.
J Biol Chem ; 286(42): 36808-19, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21880716

ABSTRACT

Galectin-1 is a member of the galectin family and has a high affinity for galactose and N-acetylglucosamine moieties of glycoproteins. It mediates multiple signal transduction pathways to modulate cellular proliferation, survival, differentiation, and migration. However, the mechanisms for the regulation of its expression remain greatly elusive. We reported previously that galectin-1 is a direct target of the hypoxia-inducible factor 1 (HIF-1), a key heterodimeric transcriptional factor for the cellular response to hypoxia. Here we show that CCAAT/enhancer binding protein α (C/EBPα), a critical transcriptional factor for hematopoietic cell differentiation, can directly activate galectin-1 through binding to the -48 to -42 bp region of its promoter. Based on the physical interaction of C/EBPα and HIF-1α, the synergistic transcriptional activity of C/EBPα and HIF-1α on the promoter of the galectin-1 gene is also found by chromatin immunoprecipitation (ChIP), ChIP followed by ChIP (ChIP-reChIP), and luciferase assay. Moreover, knockdown or chemical inhibition of galectin-1 partially blocks the differentiation induced by HIF-1α or C/EBPα, which can be rescued by recombinant galectin-1. These discoveries would shed new insights on the mechanisms for galectin-1 expression regulation and HIF-1α- and C/EBPα-induced leukemic cell differentiation.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Galectin 1/biosynthesis , Gene Expression Regulation, Leukemic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Galectin 1/genetics , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Jurkat Cells , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplasm Proteins/genetics , Response Elements/genetics , Transcription, Genetic/genetics , U937 Cells
19.
Autophagy ; 7(10): 1132-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21673516

ABSTRACT

Autophagy is a highly conserved, closely regulated homeostatic cellular activity that allows for the bulk degradation of long-lived proteins and cytoplasmic organelles. Its roles in cancer initiation and progression and in determining the response of tumor cells to anticancer therapy are complicated, and only limited investigation has been conducted on the potential significance of autophagy in the pathogenesis and therapeutic response of acute myeloid leukemia. Here we demonstrate that the inducible or transfected expression of the acute promyelocytic leukemia (APL)-specific PML-RARα, but not PLZF-RARα or NPM-RARα, fusion protein upregulates constitutive autophagy activation in leukemic and nonleukemic cells, as evaluated by hallmarks for autophagy including transmission electron microscopy. The significant increase in autophagic activity is also found in the leukemic cells-infiltrated bone marrow and spleen from PML-RARα-transplanted leukemic mice. The autophagy inhibitor 3-methyladenine significantly abrogates the autophagic events upregulated by PML-RARα, while the autophagic flux assay reveals that the fusion protein induces autophagy by increasing the on-rate of autophagic sequestration. Furthermore, this modulation of autophagy by PML-RARα is possibly mediated by a decreased activation of the Akt/mTOR pathway. Finally, we also show that autophagy contributes to the anti-apoptotic function of the PML-RARα protein. Given the critical role of the PML-RARα oncoprotein in APL pathogenesis, this study suggests an important role of autophagy in the development and treatment of this disease.


Subject(s)
Apoptosis , Autophagy , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenine/analogs & derivatives , Adenine/chemistry , Animals , Humans , Leukemia, Promyelocytic, Acute/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Neoplasm Transplantation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , TOR Serine-Threonine Kinases/antagonists & inhibitors , Time Factors , U937 Cells
20.
Carcinogenesis ; 31(8): 1367-75, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20525878

ABSTRACT

The expression of galectin-1, one of the most important lectins participating in the malignant tumor development, has been shown to be regulated by hypoxia, but its exact mechanism remains elusive. Here, we find that ectopically expressed hypoxia-inducible factor (HIF) 1alpha protein, an oxygen-sensitive subunit of HIF-1 that is a master factor for cellular response to hypoxia, significantly increases galectin-1 expression in both messenger RNA and protein levels in all four colorectal cancer (CRC) cell lines tested. However, hypoxia-induced galectin-1 expression cannot be seen in sentrin/SUMO-specific protease 1 homozygous-null mouse embryonic fibroblasts that fail to accumulate HIF-1alpha protein. Furthermore, silence of HIF-1alpha or HIF-1beta expression by specific short hairpin RNAs (shRNAs) antagonizes hypoxia-induced galectin-1 expression. All these results propose that galectin-1 is a direct target of transcriptional factor HIF-1. Applying luciferase reporter assay and chromatin immunoprecipitation, we identify that two hypoxia-responsive elements located at -441 to -423 bp upstream to transcriptional start site of galectin-1 gene are essential for HIF-1-mediated galectin-1 expression. Finally, the knockdown of galectin-1 by its specific shRNA can significantly reduce hypoxia-induced invasion and migration of CRC cell line, and the ectopic expression of galectin-1 can remarkably restore invasion and migration abilities of HIF-1alpha-knocked SW620 cells, proposing that galectin-1 mediates the HIF-1-induced migration and invasion of CRC cells during hypoxia. Taken together, our results shed new light for understanding mechanism for hypoxia/HIF-1-mediated migration/invasion of CRC cells.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Galectin 1/genetics , Hypoxia-Inducible Factor 1/physiology , Animals , Cell Line, Tumor , Cell Movement , DNA Primers , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Immunohistochemistry , Luciferases/genetics , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Plasmids , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...