1
|
Guo B, Yan S, Zhai L, Cheng Y. LncRNA HOTAIR accelerates free fatty acid-induced inflammatory response in HepG2 cells by recruiting SRSF1 to stabilize MLXIPL mRNA. Cytotechnology 2024; 76:259-269. [PMID: 38495293 PMCID: PMC10940554 DOI: 10.1007/s10616-023-00614-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 03/19/2024] Open
Abstract
LncRNA HOTAIR has been reported to be associated with metabolic diseases of the liver. However, the effect of HOTAIR on non-alcoholic fatty liver disease (NAFLD) inflammation and its potential mechanism have not been reported. Genes and proteins expression were detected by qRT-PCR and Western blot respectively. The level of inflammatory cytokines was assessed by ELISA. HepG2 cell viability was detected by MTT assay. TG level and lipid accumulation were measured by Assay Kit and Oil red O staining, respectively. Direct binding relationship between HOTAIR and Serine/arginine splicing factor 1 (SRSF1), SRSF1 and MLX interacting protein like (MLXIPL) were confirmed by RNA-pull down and RIP assay. HOTAIR was highly expressed in free fatty acids (FFA)-treated HepG2 cells. HOTAIR knockdown alleviated FFA-induced inflammation of HepG2 cells. Then further analysis showed that HOTAIR and SRSF1 had a mutual binding relationship, and HOTAIR maintained MLXIPL mRNA stability via recruiting SRSF1 in HepG2 cells. Moreover, the inhibitory effect of HOTAIR knockdown on FFA-induced inflammation in HepG2 cells was reversed by MLXIPL overexpression. HOTAIR accelerates inflammation of FFA-induced HepG2 cells by recruiting SRSF1 to stabilize MLXIPL mRNA, which will help to find new effective strategies for NAFLD therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00614-x.
Collapse
Affiliation(s)
- Bo Guo
- School of Clinical Medicine, Guangzhou Health Science College, Guangzhou, 510450 Guangdong China
| | - Shengzhe Yan
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280 Guangdong China
| | - Lei Zhai
- School of Clinical Medicine, Guangzhou Health Science College, Guangzhou, 510450 Guangdong China
| | - Yanzhen Cheng
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280 Guangdong China
| |
Collapse
|
2
|
Benichou E, Seffou B, Topçu S, Renoult O, Lenoir V, Planchais J, Bonner C, Postic C, Prip-Buus C, Pecqueur C, Guilmeau S, Alves-Guerra MC, Dentin R. The transcription factor ChREBP Orchestrates liver carcinogenesis by coordinating the PI3K/AKT signaling and cancer metabolism. Nat Commun 2024; 15:1879. [PMID: 38424041 PMCID: PMC10904844 DOI: 10.1038/s41467-024-45548-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Cancer cells integrate multiple biosynthetic demands to drive unrestricted proliferation. How these cellular processes crosstalk to fuel cancer cell growth is still not fully understood. Here, we uncover the mechanisms by which the transcription factor Carbohydrate responsive element binding protein (ChREBP) functions as an oncogene during hepatocellular carcinoma (HCC) development. Mechanistically, ChREBP triggers the expression of the PI3K regulatory subunit p85α, to sustain the activity of the pro-oncogenic PI3K/AKT signaling pathway in HCC. In parallel, increased ChREBP activity reroutes glucose and glutamine metabolic fluxes into fatty acid and nucleic acid synthesis to support PI3K/AKT-mediated HCC growth. Thus, HCC cells have a ChREBP-driven circuitry that ensures balanced coordination between PI3K/AKT signaling and appropriate cell anabolism to support HCC development. Finally, pharmacological inhibition of ChREBP by SBI-993 significantly suppresses in vivo HCC tumor growth. Overall, we show that targeting ChREBP with specific inhibitors provides an attractive therapeutic window for HCC treatment.
Collapse
Affiliation(s)
- Emmanuel Benichou
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Bolaji Seffou
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Selin Topçu
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Ophélie Renoult
- Nantes Université, INSERM U1307, CNRS 6075, CRCI2NA, Nantes, France
| | - Véronique Lenoir
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Julien Planchais
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Caroline Bonner
- Institut Pasteur de Lille, Lille, France
- INSERM, U1011, Lille, France
- European Genomic Institute for Diabetes, Lille, France
- Université de Lille, Lille, France
| | - Catherine Postic
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Carina Prip-Buus
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Claire Pecqueur
- Nantes Université, INSERM U1307, CNRS 6075, CRCI2NA, Nantes, France
| | - Sandra Guilmeau
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | | | - Renaud Dentin
- Université Paris Cité, Institut Cochin, INSERM, CNRS, F-75014, Paris, France.
- Institut Cochin, Faculté de Médecine 3ème étage, 24 Rue du Faubourg Saint Jacques, 75014, Paris, France.
| |
Collapse
|
3
|
Chen J, Chan TTH, Zhou J. Lipid metabolism in the immune niche of tumor-prone liver microenvironment. J Leukoc Biol 2024; 115:68-84. [PMID: 37474318 DOI: 10.1093/jleuko/qiad081] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/23/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
The liver is a common primary site not only for tumorigenesis, but also for cancer metastasis. Advanced cancer patients with liver metastases also show reduced response rates and survival benefits when treated with immune checkpoint inhibitors. Accumulating evidence has highlighted the importance of the liver immune microenvironment in determining tumorigenesis, metastasis-organotropism, and immunotherapy resistance. Various immune cells such as T cells, natural killer and natural killer T cells, macrophages and dendritic cells, and stromal cells including liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, and hepatocytes are implicated in contributing to the immune niche of tumor-prone liver microenvironment. In parallel, as the major organ for lipid metabolism, the increased abundance of lipids and their metabolites is linked to processes crucial for nonalcoholic fatty liver disease and related liver cancer development. Furthermore, the proliferation, differentiation, and functions of hepatic immune and stromal cells are also reported to be regulated by lipid metabolism. Therefore, targeting lipid metabolism may hold great potential to reprogram the immunosuppressive liver microenvironment and synergistically enhance the immunotherapy efficacy in the circumstance of liver metastasis. In this review, we describe how the hepatic microenvironment adapts to the lipid metabolic alterations in pathologic conditions like nonalcoholic fatty liver disease. We also illustrate how these immunometabolic alterations promote the development of liver cancers and immunotherapy resistance. Finally, we discuss the current therapeutic options and hypothetic combination immunotherapies for the treatment of advanced liver cancers.
Collapse
Affiliation(s)
- Jintian Chen
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Thomas T H Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Jingying Zhou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| |
Collapse
|
4
|
Santamarina AB, Mennitti LV, de Souza EA, Mesquita LMDS, Noronha IH, Vasconcelos JRC, Prado CM, Pisani LP. A low-carbohydrate diet with different fatty acids' sources in the treatment of obesity: Impact on insulin resistance and adipogenesis. Clin Nutr 2023; 42:2381-2394. [PMID: 37862824 DOI: 10.1016/j.clnu.2023.09.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/01/2023] [Accepted: 09/23/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND The search for nutritional intervention strategies against obesity has grown, highlighting the low-carbohydrate diet model. However, little is known about the impact of the quality of fatty acids consumed in this diet. Thus, we aim to investigate the influence of fatty acid quality on dietary strategy on obesity. METHODS Male Swiss mice were diet-induced to obesity. Afterward, mice consume a low-carb diet with different types of fat: saturated, polyunsaturated ω-3, ω-6, and monounsaturated ω-9 fatty acids. Weight gain and food consumption were monitored weekly. An oral glucose tolerance test was performed and blood and tissue samples were collected for analysis of insulin resistance markers. Protein expression of insulin signaling pathway molecules, lipid metabolism, mitochondrial function, macrophage polarization, and cytokine production were analyzed. RESULTS The high-fat diet was able to induce obesity and glucose intolerance. The switch to a low-carbohydrate dietary pattern reversed the glucose intolerance, with better results in the ω-3 and ω-9 groups. After the low-carbohydrate diet, groups ω-3 and ω-9 presented improved fasting serum glucose, insulin, and HOMA indexes. The low-carbohydrate diet also increased the activity of insulin pathway proteins such as IR, IRS1, and AKT. Furthermore, the ω-3 diet group showed greater activity of mitochondrial complexes and AMPK signaling pathway proteins. The ω-6 and ω-9 -rich diet induced M2-type macrophage polarization, as well as cytokine production modulation by the low-carbohydrate diet in the ω-3 and ω-9 groups. CONCLUSIONS Consuming a low-carbohydrate diet pattern promotes weight loss and improves glucose intolerance in obesity. Also, the quality of lipids has a direct influence, demonstrating that the consumption of ω-3 polyunsaturated and ω-9 monounsaturated lipids can lead to more favorable outcomes for the improvement of glucose intolerance, lipid metabolism, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Aline B Santamarina
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Laís V Mennitti
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Esther A de Souza
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Leonardo M de Souza Mesquita
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas, Rua Pedro Zaccaria 1300, 13484-350 Limeira, São Paulo, Brazil
| | - Isaú H Noronha
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - José Ronnie C Vasconcelos
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Carla M Prado
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo, Campus Baixada Santista - UNIFESP, Santos, São Paulo, Brazil.
| |
Collapse
|
5
|
Chen M, Lin Y, Dang Y, Xiao Y, Zhang F, Sun G, Jiang X, Zhang L, Du J, Duan S, Zhang X, Qin Z, Yang J, Liu K, Wu B. Reprogramming of rhythmic liver metabolism by intestinal clock. J Hepatol 2023; 79:741-757. [PMID: 37230230 DOI: 10.1016/j.jhep.2023.04.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND & AIMS Temporal oscillations in intestinal nutrient processing and absorption are coordinated by the local clock, which leads to the hypothesis that the intestinal clock has major impacts on shaping peripheral rhythms via diurnal nutritional signals. Here, we investigate the role of the intestinal clock in controlling liver rhythmicity and metabolism. METHODS Transcriptomic analysis, metabolomics, metabolic assays, histology, quantitative (q)PCR, and immunoblotting were performed with Bmal1-intestine-specific knockout (iKO), Rev-erba-iKO, and control mice. RESULTS Bmal1 iKO caused large-scale reprogramming of the rhythmic transcriptome of mouse liver with a limited effect on its clock. In the absence of intestinal Bmal1, the liver clock was resistant to entrainment by inverted feeding and a high-fat diet. Importantly, Bmal1 iKO remodelled diurnal hepatic metabolism by shifting to gluconeogenesis from lipogenesis during the dark phase, leading to elevated glucose production (hyperglycaemia) and insulin insensitivity. Conversely, Rev-erba iKO caused a diversion to lipogenesis from gluconeogenesis during the light phase, resulting in enhanced lipogenesis and an increased susceptibility to alcohol-related liver injury. These temporal diversions were attributed to disruption of hepatic SREBP-1c rhythmicity, which was maintained via gut-derived polyunsaturated fatty acids produced by intestinal FADS1/2 under the control of a local clock. CONCLUSIONS Our findings establish a pivotal role for the intestinal clock in dictating liver rhythmicity and diurnal metabolism, and suggest targeting intestinal rhythms as a new avenue for improving metabolic health. IMPACT AND IMPLICATIONS Our findings establish the centrality of the intestinal clock among peripheral tissue clocks, and associate liver-related pathologies with its malfunction. Clock modifiers in the intestine are shown to modulate liver metabolism with improved metabolic parameters. Such knowledge will help clinicians improve the diagnosis and treatment of metabolic diseases by incorporating intestinal circadian factors.
Collapse
Affiliation(s)
- Min Chen
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanke Lin
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongkang Dang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifei Xiao
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fugui Zhang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guanghui Sun
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuejun Jiang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Zhang
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhao Du
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuyi Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zifei Qin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kaisheng Liu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
6
|
Gugliucci A. Sugar and Dyslipidemia: A Double-Hit, Perfect Storm. J Clin Med 2023; 12:5660. [PMID: 37685728 PMCID: PMC10488931 DOI: 10.3390/jcm12175660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/10/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
The availability of sugar has expanded over the past 50 years, due to improved industrial processes and corn subsidies, particularly in the form of sweetened beverages. This correlates with a surge in the prevalence of cardiometabolic disorders, which has brought this issue back into the spotlight for public health. In this narrative review, we focus on the role of fructose in the genesis of cardiometabolic dyslipidemia (an increase in serum triglyceride-rich lipoproteins (TRL): VLDL, chylomicrons (CM), and their remnants) bringing together the most recent data on humans, which demonstrates the crucial interaction between glucose and fructose, increasing the synthesis while decreasing the catabolism of these particles in a synergistic downward spiral. After reviewing TRL metabolism, we discuss the fundamental principles governing the metabolism of fructose in the intestine and liver and the effects of dysregulated fructolysis, in conjunction with the activation of carbohydrate-responsive element-binding protein (ChREBP) by glucose and the resulting crosstalk. The first byproduct of fructose catabolism, fructose-1-P, is highlighted for its function as a signaling molecule that promotes fat synthesis. We emphasize the role of fructose/glucose interaction in the liver, which enhances de novo lipogenesis, triglyceride (TG) synthesis, and VLDL production. In addition, we draw attention to current research that demonstrates how fructose affects the activity of lipoprotein lipase by increasing the concentration of inhibitors such as apolipoprotein CIII (apoCIII) and angiopoietin-like protein 3 (ANGPTL3), which reduce the catabolism of VLDL and chylomicrons and cause the building up of their atherogenic remnants. The end outcome is a dual, synergistic, and harmful action that encourages atherogenesis. Thus, considering the growing concerns regarding the connection between sugar consumption and cardiometabolic disease, current research strongly supports the actions of public health organizations aimed at reducing sugar intake, including dietary guidance addressing "safe" limits for sugar consumption.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
7
|
Inhibition of ChREBP ubiquitination via the ROS/Akt-dependent downregulation of Smurf2 contributes to lysophosphatidic acid-induced fibrosis in renal mesangial cells. J Biomed Sci 2022; 29:31. [PMID: 35538534 PMCID: PMC9092836 DOI: 10.1186/s12929-022-00814-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/29/2022] [Indexed: 12/20/2022] Open
Abstract
Background Mesangial cell fibrosis, a typical symptom of diabetic nephropathy (DN), is a major contributor to glomerulosclerosis. We previously reported that the pharmacological blockade of lysophosphatidic acid (LPA) signaling improves DN. Although LPA signaling is implicated in diabetic renal fibrosis, the underlying molecular mechanisms remain unclear. Here, the role of carbohydrate-responsive element-binding protein (ChREBP) in LPA-induced renal fibrosis and the underlying mechanisms were investigated. Methods Eight-week-old wild-type and db/db mice were intraperitoneally injected with the vehicle or an LPAR1/3 antagonist, ki16425 (10 mg/kg), for 8 weeks on a daily basis, following which the mice were sacrificed and renal protein expression was analyzed. SV40 MES13 cells were treated with LPA in the presence or absence of ki16425, and the expression of ChREBP and fibrotic factors, including fibronectin, TGF-β, and IL-1β, was examined. The role of ChREBP in the LPA-induced fibrotic response was investigated by ChREBP overexpression or knockdown. The involvement of Smad ubiquitination regulatory factor-2 (Smurf2), an E3 ligase, in LPA-induced expression of ChREBP and fibrotic factors was investigated by Smurf2 overexpression or knockdown. To identify signaling molecules regulating Smurf2 expression by LPA, pharmacological inhibitors such as A6370 (Akt1/2 kinase inhibitor) and Ly 294002 (PI3K inhibitor) were used. Results The renal expression of ChREBP increased in diabetic db/db mice, and was reduced following treatment with the ki16425. Treatment with LPA induced the expression of ChREBP and fibrotic factors, including fibronectin, TGF-β, and IL-1β, in SV40 MES13 cells, which were positively correlated. The LPA-induced expression of fibrotic factors increased or decreased following ChREBP overexpression and knockdown, respectively. The production of reactive oxygen species (ROS) mediated the LPA-induced expression of ChREBP and fibrotic factors, and LPA decreased Smurf2 expression via Traf4-mediated ubiquitination. The LPA-induced expression of ubiquitinated-ChREBP increased or decreased following Smurf2 overexpression and knockdown, respectively. Additionally, Smurf2 knockdown significantly increased the expression of ChREBP and fibrotic factors. The pharmacological inhibition of Akt signaling suppressed the LPA-induced alterations in the expression of ChREBP and Smurf2. Conclusion Collectively, the results demonstrated that the ROS/Akt-dependent downregulation of Smurf2 and the subsequent increase in ChREBP expression might be one of the mechanisms by which LPA induces mesangial cell fibrosis in DN. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00814-1.
Collapse
|
8
|
Abstract
Mammals undergo regular cycles of fasting and feeding that engage dynamic transcriptional responses in metabolic tissues. Here we review advances in our understanding of the gene regulatory networks that contribute to hepatic responses to fasting and feeding. The advent of sequencing and -omics techniques have begun to facilitate a holistic understanding of the transcriptional landscape and its plasticity. We highlight transcription factors, their cofactors, and the pathways that they impact. We also discuss physiological factors that impinge on these responses, including circadian rhythms and sex differences. Finally, we review how dietary modifications modulate hepatic gene expression programs.
Collapse
Affiliation(s)
- Lara Bideyan
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Rohith Nagari
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
9
|
Płatek T, Polus A, Góralska J, Raźny U, Dziewońska A, Micek A, Dembińska-Kieć A, Solnica B, Malczewska-Malec M. Epigenetic Regulation of Processes Related to High Level of Fibroblast Growth Factor 21 in Obese Subjects. Genes (Basel) 2021; 12:307. [PMID: 33670024 PMCID: PMC7926457 DOI: 10.3390/genes12020307] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
We hypothesised that epigenetics may play an important role in mediating fibroblast growth factor 21 (FGF21) resistance in obesity. We aimed to evaluate DNA methylation changes and miRNA pattern in obese subjects associated with high serum FGF21 levels. The study included 136 participants with BMI 27-45 kg/m2. Fasting FGF21, glucose, insulin, GIP, lipids, adipokines, miokines and cytokines were measured and compared in high serum FGF21 (n = 68) group to low FGF21 (n = 68) group. Human DNA Methylation Microarrays were analysed in leukocytes from each group (n = 16). Expression of miRNAs was evaluated using quantitative PCR-TLDA. The study identified differentially methylated genes in pathways related to glucose transport, insulin secretion and signalling, lipid transport and cellular metabolism, response to nutrient levels, thermogenesis, browning of adipose tissue and bone mineralisation. Additionally, it detected transcription factor genes regulating FGF21 and fibroblast growth factor receptor and vascular endothelial growth factor receptor pathways regulation. Increased expression of hsa-miR-875-5p and decreased expression of hsa-miR-133a-3p, hsa-miR-185-5p and hsa-miR-200c-3p were found in the group with high serum FGF21. These changes were associated with high FGF21, VEGF and low adiponectin serum levels. Our results point to a significant role of the epigenetic regulation of genes involved in metabolic pathways related to FGF21 action.
Collapse
Affiliation(s)
- Teresa Płatek
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Anna Polus
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Joanna Góralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Urszula Raźny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Agnieszka Dziewońska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Agnieszka Micek
- Department of Nursing Management and Epidemiology Nursing, Faculty of Health Sciences, Jagiellonian University Medical College, 25 Kopernika Street, 31-501 Krakow, Poland;
| | - Aldona Dembińska-Kieć
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Bogdan Solnica
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| | - Małgorzata Malczewska-Malec
- Department of Clinical Biochemistry, Jagiellonian University Medical College, 15a Kopernika Street, 31-501 Krakow, Poland; (A.P.); (J.G.); (U.R.); (A.D.); (A.D.-K.); (B.S.); (M.M.-M.)
| |
Collapse
|
10
|
Ozkan H, Yakan A. Dietary high calories from sunflower oil, sucrose and fructose sources alters lipogenic genes expression levels in liver and skeletal muscle in rats. Ann Hepatol 2020; 18:715-724. [PMID: 31204236 DOI: 10.1016/j.aohep.2019.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/25/2019] [Accepted: 03/19/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES The objectives of this study were to investigate the underlying mechanism of PPARα, LXRα, ChREBP, and SREBP-1c at the level of gene and protein expression with high-energy diets in liver and skeletal muscle. MATERIALS AND METHODS Metabolic changes with consumption of high fat (Hfat), high sucrose (Hsuc) and high fructose (Hfru) diets were assessed. Levels of mRNA and protein of PPARα, LXRα, ChREBP, and SREBP-1c were investigated. Body weight changes, histological structure of liver and plasma levels of some parameters were also examined. RESULTS In Hfru group, body weights were higher than other groups (P<0.05). In liver, LXRα levels of Hsuc and Hfru groups were upregulated as 1.87±0.30 (P<0.05) and 2.01±0.29 (P<0.01). SREBP-1c levels were upregulated as 4.52±1.25 (P<0.05); 4.05±1.11 (P<0.05) and 3.85±1.04 (P<0.05) in Hfat, Hsuc, and Hfru groups, respectively. In skeletal muscle, LXRα and SREBP-1c were upregulated as 1.77±0.30 (P<0.05) and 2.71±0.56 (P<0.05), in the Hfru group. Protein levels of ChREBP (33.92±8.84ng/mg protein (P<0.05)) and SREBP-1c (135.16±15.57ng/mg protein (P<0.001)) in liver were higher in Hfru group. In skeletal muscle, LXRα, ChREBP and SREBP-1c in Hfru group were 6.67±0.60, 7.11±1.29 and 43.17±6.37ng/mg, respectively (P<0.05; P<0.01; P<0.05). The rats in Hfru group had the most damaged livers. CONCLUSION Besides liver, fructose consumption significantly effects skeletal muscle and leads to weight gain, triggers lipogenesis and metabolic disorders.
Collapse
Affiliation(s)
- Huseyin Ozkan
- Department of Genetic, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, Turkey.
| | - Akin Yakan
- Department of Animal Breeding, Faculty of Veterinary Medicine, University of Erciyes, Kayseri, Turkey
| |
Collapse
|
11
|
Tu G, Dai C, Qu H, Wang Y, Liao B. Role of exercise and rapamycin on the expression of energy metabolism genes in liver tissues of rats fed a high‑fat diet. Mol Med Rep 2020; 22:2932-2940. [PMID: 32945385 PMCID: PMC7453655 DOI: 10.3892/mmr.2020.11362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 06/26/2020] [Indexed: 12/19/2022] Open
Abstract
The mTOR pathway serves an important role in the development of insulin resistance induced by obesity. Exercise improves obesity-associated insulin resistance and hepatic energy metabolism; however, the precise mechanism of this process remains unknown. Therefore, the present study investigated the role of rapamycin, an inhibitor of mTOR, on exercise-induced expression of hepatic energy metabolism genes in rats fed a high-fat diet (HFD). A total of 30 male rats were divided into the following groups: Normal group (n=6) fed chow diets and HFD group (n=24) fed an HFD for 6 weeks. The HFD rats performed exercise adaptation for 1 week and were randomly divided into the four following groups (each containing six rats): i) Group of HFD rats with sedentary (H group); ii) group of HFD rats with exercise (HE group); iii) group of HFD rats with rapamycin (HR group); and iv) group of HFD rats with exercise and rapamycin (HER group). Both HE and HER rats were placed on incremental treadmill training for 4 weeks (from week 8–11). Both HR and HER rats were injected with rapamycin intraperitoneally at the dose of 2 mg/kg once a day for 2 weeks (from week 10–11). All rats were sacrificed following a 12–16 h fasting period at the end of week 11. The levels of mitochondrial and oxidative enzyme activities, as well as of the expression of genes involved in energy metabolism were assessed in liver tissues. Biochemical assays and oil red staining were used to assess the content of hepatic triglycerides (TGs). The results indicated that exercise, but not rapamycin, reduced TG content in the liver of HFD rats. Further analysis indicated that rapamycin reduced the activity of cytochrome c oxidase, but not the activities of succinate dehydrogenase and β-hydroxyacyl-CoA dehydrogenase in the liver of HFD rats. Exercise significantly upregulated the mRNA expression of peroxisome proliferator-activated receptor γ coactivator 1 β, while rapamycin exhibited no effect on the mRNA expression levels of hepatic transcription factors associated with energy metabolism enzymes in the liver of HFD rats. Collectively, the results indicated that exercise reduced TG content and upregulated mitochondrial metabolic gene expression in the liver of HFD rats. Moreover, this mechanism may not involve the mTOR pathway.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Chunyan Dai
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Haofei Qu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Yunzhen Wang
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| |
Collapse
|
12
|
Pardo PS, Boriek AM. SIRT1 Regulation in Ageing and Obesity. Mech Ageing Dev 2020; 188:111249. [PMID: 32320732 DOI: 10.1016/j.mad.2020.111249] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 03/12/2020] [Accepted: 04/05/2020] [Indexed: 12/29/2022]
Abstract
Ageing and obesity have common hallmarks: altered glucose and lipid metabolism, chronic inflammation and oxidative stress are some examples. The downstream effects of SIRT1 activity have been thoroughly explored, and their research is still in expanse. SIRT1 activation has been shown to regulate pathways with beneficiary effects on 1) ageing and obesity-associated metabolic disorders such as metabolic syndrome, insulin resistance and type-II diabetes with, 2) chronic inflammatory processes such as arthritis, atherosclerosis and emphysema, 3) DNA damage and oxidative stress with impact on neurodegenerative diseases, cardiovascular health and some cancers. This knowledge intensified the interest in uncovering the mechanisms regulating the expression and activity of SIRT1. This review focuses on the upstream regulatory mechanisms controlling SIRT1, and how this knowledge could potentially contribute to the development of therapeutic interventions.
Collapse
Affiliation(s)
- Patricia S Pardo
- Pulmonary and Critical Care medicine, Department of Medicine, Baylor College of Medicine, Houston TX 77030, USA.
| | - Aladin M Boriek
- Pulmonary and Critical Care medicine, Department of Medicine, Baylor College of Medicine, Houston TX 77030, USA.
| |
Collapse
|
13
|
Al-Attar R, Storey KB. Suspended in time: Molecular responses to hibernation also promote longevity. Exp Gerontol 2020; 134:110889. [PMID: 32114078 DOI: 10.1016/j.exger.2020.110889] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022]
Abstract
Aging in most animals is an inevitable process that causes or is a result of physiological, biochemical, and molecular changes in the body, and has a strong influence on an organism's lifespan. Although advancement in medicine has allowed humans to live longer, the prevalence of age-associated medical complications is continuously burdening older adults worldwide. Current animal models used in research to study aging have provided novel information that has helped investigators understand the aging process; however, these models are limiting. Aging is a complex process that is regulated at multiple biological levels, and while a single manipulation in these models can provide information on a process, it is not enough to understand the global regulation of aging. Some mammalian hibernators live up to 9.8-times higher than their expected average lifespan, and new research attributes this increase to their ability to hibernate. A common theme amongst these mammalian hibernators is their ability to greatly reduce their metabolic rate to a fraction of their normal rate and initiate cytoprotective responses that enable their survival. Metabolic rate depression is strictly regulated at different biological levels in order to enable the animal to not only survive, but to also do so by relying mainly on their limited internal fuels. As such, understanding both the global and specific regulatory mechanisms used to promote survival during hibernation could, in theory, allow investigators to have a better understanding of the aging process. This can also allow pharmaceutical industries to find therapeutics that could delay or reverse age-associated medical complications and promote healthy aging and longevity in humans.
Collapse
Affiliation(s)
- Rasha Al-Attar
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | - Kenneth B Storey
- Institute of Biochemistry and Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| |
Collapse
|
14
|
Spalletta S, Flati V, Toniato E, Di Gregorio J, Marino A, Pierdomenico L, Marchisio M, D’Orazi G, Cacciatore I, Robuffo I. Carvacrol reduces adipogenic differentiation by modulating autophagy and ChREBP expression. PLoS One 2018; 13:e0206894. [PMID: 30418986 PMCID: PMC6231630 DOI: 10.1371/journal.pone.0206894] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 10/22/2018] [Indexed: 01/09/2023] Open
Abstract
Objective Obesity is the result of white adipose tissue accumulation where excess of food energy is stored to form triglycerides. De novo lipogenesis (DNL) is the continuous process of new fat production and is driven by the transcription factor ChREBP. During adipogenesis, white adipocytes change their morphology and the entire cell volume is occupied by one large lipid droplet. Recent studies have implicated an essential role of autophagy in adipogenic differentiation, cytoplasmic remodelling and mitochondria reorganization. The phenolic monoterpenoid carvacrol (2-methyl-5-[1-methylethyl]phenol), produced by numerous aromatic plants, has been shown to reduce lipid accumulation in murine 3T3-L1 cells during adipogenic differentiation by modulating genes associated with adipogenesis and inflammation. Therefore, the aim of this study was to evaluate whether carvacrol could affect autophagy and ChREBP expression during adipogenic differentiation. Methods The study was carried on by using the murine 3T3-L1 and the human WJ-MSCs (Wharton’s jelly-derived mesenchymal stem cells) cell lines. Cells undergoing adipogenic differentiation were untreated or treated with carvacrol. Adipogenic differentiation was assessed by analyzing cellular lipid accumulation with Oil-Red O staining and by ultrastructural examination with TEM. Autophagy was evaluated by western immunoblotting of autophagy markers LC3B and p62/SQSTM and by ultrastructural examination of autophagic bodies. Autophagic flux was evaluated by using autophagy inhibitor cloroquine (CQ). ChREBP expression levels was assessed by both western blotting and immunoelectron microscopy and ChREBP activity by analysis of adipogenic target genes expression. Results We found that carvacrol reduced adipogenic differentiation of about 40% and 30% in, respectively, 3T3-L1 and in WJ-MSCs cells. The effect of carvacrol on adipogenic differentiation correlated with both reduction of autophagy and reduction of ChREBP expression. Conclusion The results support the notion that carvacrol, through its effect on autophagy (essential for adipocyte maturation) and on ChREBP activity, could be used as a valuable adjuvant to reduce adipogenic differentiation.
Collapse
Affiliation(s)
- Sonia Spalletta
- Institute of Molecular Genetics, National Research Council, Section of Chieti, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Elena Toniato
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Jacopo Di Gregorio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Antonio Marino
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Laura Pierdomenico
- Department of Medicine and Aging Science, University “G. d’Annunzio”, Chieti-Pescara, Chieti, Italy
- Aging Research Center (CeSI-Met), Gabriele D’Annunzio University Foundation, Chieti, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Science, University “G. d’Annunzio”, Chieti-Pescara, Chieti, Italy
- Aging Research Center (CeSI-Met), Gabriele D’Annunzio University Foundation, Chieti, Italy
| | - Gabriella D’Orazi
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Iole Robuffo
- Institute of Molecular Genetics, National Research Council, Section of Chieti, Chieti, Italy
- Department of Medicine and Aging Science, University “G. d’Annunzio”, Chieti-Pescara, Chieti, Italy
- * E-mail:
| |
Collapse
|
15
|
Libby AE, Bales ES, Monks J, Orlicky DJ, McManaman JL. Perilipin-2 deletion promotes carbohydrate-mediated browning of white adipose tissue at ambient temperature. J Lipid Res 2018; 59:1482-1500. [PMID: 29866659 DOI: 10.1194/jlr.m086249] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/31/2018] [Indexed: 12/22/2022] Open
Abstract
Mice lacking perilipin-2 (Plin2-null) are resistant to obesity, insulin resistance, and fatty liver induced by Western or high-fat diets. In the current study, we found that, compared with WT mice on Western diet, Plin2-null adipose tissue was more insulin sensitive and inguinal subcutaneous white adipose tissue (iWAT) exhibited profound browning and robust induction of thermogenic and carbohydrate-responsive genetic programs at room temperature. Surprisingly, these Plin2-null responses correlated with the content of simple carbohydrates, rather than fat, in the diet, and were independent of adipose Plin2 expression. To define Plin2 and sugar effects on adipose browning, WT and Plin2-null mice were placed on chow diets containing 20% sucrose in their drinking water for 6 weeks. Compared with WT mice, iWAT of Plin2-null mice exhibited pronounced browning and striking increases in the expression of thermogenic and insulin-responsive genes on this diet. Significantly, Plin2-null iWAT browning was associated with reduced sucrose intake and elevated serum fibroblast growth factor (FGF)21 levels, which correlated with greatly enhanced hepatic FGF21 production. These data identify Plin2 actions as novel mediators of sugar-induced adipose browning through indirect effects of hepatic FGF21 expression, and suggest that adipose browning mechanisms may contribute to Plin2-null resistance to obesity.
Collapse
Affiliation(s)
- Andrew E Libby
- Integrated Physiology Graduate Program, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045.,Division of Reproductive Sciences, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045
| | - Elise S Bales
- Division of Reproductive Sciences, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045
| | - Jenifer Monks
- Division of Reproductive Sciences, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045
| | - David J Orlicky
- Department of Pathology, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045
| | - James L McManaman
- Integrated Physiology Graduate Program, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045 .,Division of Reproductive Sciences, University of Colorado at Denver, Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
16
|
Cai C, Yu H, Huang G, Du X, Yu X, Zhou Y, Shen W. Histone modifications in fatty acid synthase modulated by carbohydrate responsive element binding protein are associated with non‑alcoholic fatty liver disease. Int J Mol Med 2018; 42:1215-1228. [PMID: 29786745 PMCID: PMC6089769 DOI: 10.3892/ijmm.2018.3702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a manifestation of metabolic syndrome in the liver and is closely associated with diabetes; however, its pathogenesis remains to be elucidated. Carbohydrate responsive element binding protein (ChREBP), the hub of glucolipid metabolism, regulates the induction of fatty acid synthase (FASN), the key enzyme of de novo lipogenesis, by directly binding to carbohydrate response element (ChoRE) in its promoter. Investigations of histone modifications on NAFLD remain in their infancy. In the present study, by using ChIP, the association between histone modifications and FASN transcription was investigated and histone modifications in FASN modulated by ChREBP were measured. It was demonstrated that ChREBP induced FASN ChREBP-ChoRE binding to accelerate the expression of FASN, leading to hepatocellular steatosis by facilitating H3 and H4 acetylation, H3K4 trimethylation and the phosphorylation of H3S10, but inhibiting the trimethylation of H3K9 and H4K20 in FASN promoter regions of HepG2 and L02 cells. It was also found that ChREBP-ChoRE binding of FASN relied on histone acetylation and that the transcriptional activity of ChREBP on FASN is required, based on the premise that histone acetylation causes conformational changes in FASN chromatin. This indicated histone acetylation as a crucial mechanism involved in the transcription of FASN modulated by ChREBP. Consequently, the present study provides further insight into the pathophysiology and a novel therapeutic potential of NAFLD based on epigenetic mechanisms.
Collapse
Affiliation(s)
- Can Cai
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Huihong Yu
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Guangming Huang
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xuan Du
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xiaoqing Yu
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Youping Zhou
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Wei Shen
- Department of Gastroenterology, 2nd Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
17
|
Zhang X, Yang W, Wang J, Meng Y, Guan Y, Yang J. FAM3 gene family: A promising therapeutical target for NAFLD and type 2 diabetes. Metabolism 2018; 81:71-82. [PMID: 29221790 DOI: 10.1016/j.metabol.2017.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/08/2017] [Accepted: 12/01/2017] [Indexed: 12/15/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and diabetes are severe public health issues worldwide. The Family with sequence similarity 3 (FAM3) gene family consists of four members designated as FAM3A, FAM3B, FAM3C and FAM3D, respectively. Recently, there had been increasing evidence that FAM3A, FAM3B and FAM3C are important regulators of glucose and lipid metabolism. FAM3A expression is reduced in the livers of diabetic rodents and NAFLD patients. Hepatic FAM3A restoration activates ATP-P2 receptor-Akt and AMPK pathways to attenuate steatosis and hyperglycemia in obese diabetic mice. FAM3C expression is also reduced in the liver under diabetic condition. FAM3C is a new hepatokine that activates HSF1-CaM-Akt pathway and represses mTOR-SREBP1-FAS pathway to suppress hepatic gluconeogenesis and lipogenesis. In contrast, hepatic expression of FAM3B, also called PANDER, is increased under obese state. FAM3B promotes hepatic lipogenesis and gluconeogenesis by repressing Akt and AMPK activities, and activating lipogenic pathway. Under obese state, the imbalance among hepatic FAM3A, FAM3B and FAM3C signaling networks plays important roles in the pathogenesis of NAFLD and type 2 diabetes. This review briefly discussed the latest research progress on the roles and mechanisms of FAM3A, FAM3B and FAM3C in the regulation of hepatic glucose and lipid metabolism.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Weili Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Junpei Wang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Yuhong Meng
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Jichun Yang
- Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
18
|
Jois T, Sleeman MW. The regulation and role of carbohydrate response element-binding protein in metabolic homeostasis and disease. J Neuroendocrinol 2017; 29. [PMID: 28370553 DOI: 10.1111/jne.12473] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/26/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
The transcription factor carbohydrate response element-binding protein (ChREBP) is a member of the basic helix-loop-helix leucine zipper transcription factor family. Under high-glucose conditions, it has a role in regulating the expression of key genes involved in various pathways, including glycolysis, gluconeogenesis and lipogenesis. It does this by forming a tetrameric complex made up of two ChREBP/Mlx heterodimers, which enables it to bind to the carbohydrate response element (ChoRE) in the promoter region of its target genes to regulate transcription. Because ChREBP plays a key role in glucose signalling and metabolism, and aberrations in glucose homeostasis are often present in metabolic diseases, this transcription factor presents itself as an enticing target with respect to further understanding metabolic disease mechanisms and potentially uncovering new therapeutic targets.
Collapse
Affiliation(s)
- T Jois
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M W Sleeman
- Department of Physiology, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
19
|
Soares CT, Trombone APF, Fachin LRV, Rosa PS, Ghidella CC, Ramalho RF, Pinilla MG, Carvalho AF, Carrara DN, Soares FA, Belone AFF. Differential Expression of MicroRNAs in Leprosy Skin Lesions. Front Immunol 2017; 8:1035. [PMID: 28970833 PMCID: PMC5609578 DOI: 10.3389/fimmu.2017.01035] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 08/10/2017] [Indexed: 12/19/2022] Open
Abstract
Leprosy, a chronic infectious disease caused by Mycobacterium leprae, is a major public health problem in poor and developing countries of the Americas, Africa, and Asia. MicroRNAs (miRNAs), which are small non-coding RNAs (18–24 nucleotides), play an important role in regulating cell and tissue homeostasis through translational downregulation of messenger RNAs (mRNAs). Deregulation of miRNA expression is important for the pathogenesis of various neoplastic and non-neoplastic diseases and has been the focus of many publications; however, studies on the expression of miRNAs in leprosy are rare. Herein, an extensive evaluation of differentially expressed miRNAs was performed on leprosy skin lesions using microarrays. Leprosy patients, classified according to Ridley and Jopling’s classification or reactional states (R1 and R2), and healthy controls (HCs) were included. Punch biopsies were collected from the borders of leprosy lesions (10 tuberculoid, 10 borderline tuberculoid, 10 borderline borderline, 10 borderline lepromatous, 4 lepromatous, 14 R1, and 9 R2) and from 9 HCs. miRNA expression profiles were obtained using the Agilent Microarray platform with miRBase, which consists of 1,368 Homo sapiens (hsa)-miRNA candidates. TaqMan quantitative real-time reverse transcription polymerase chain reaction (RT-PCR) was used to validate differentially expressed miRNAs. Sixty-four differentially expressed miRNAs, including 50 upregulated and 14 downregulated (fold change ≥2.0, p-value ≤ 0.05) were identified after comparing samples from patients to those of controls. Twenty differentially expressed miRNAs were identified exclusively in the reactional samples (14 type 1 and 6 type 2). Eight miRNAs were validated by RT-PCR, including seven upregulated (hsa-miR-142-3p, hsa-miR-142-5p, hsa-miR-146b-5p, hsa-miR-342-3p, hsa-miR-361-3p, hsa-miR-3653, and hsa-miR-484) and one downregulated (hsa-miR-1290). These miRNAs were differentially expressed in leprosy and several other diseases, especially those related to the immune response. Moreover, the integration of analysis of validated mi/mRNAs obtained from the same samples allowed target pairs opposite expression pattern of hsa-miRNA-142-3p and AKR1B10, hsa-miRNA-342-3p and FAM180b, and hsa-miRNA-484 and FASN. This study identified several miRNAs that might play an important role in the molecular pathogenesis of the disease. Moreover, these deregulated miRNAs and their respective signaling pathways might be useful as therapeutic markers, therapeutic targets, which could help in the development of drugs to treat leprosy.
Collapse
Affiliation(s)
- Cleverson T Soares
- Department of Anatomic Pathology, Instituto Lauro de Souza Lima, São Paulo, Brazil
| | - Ana P F Trombone
- Department of Health Science, Universidade do Sagrado Coração, São Paulo, Brazil
| | - Luciana R V Fachin
- Department of Anatomic Pathology, Instituto Lauro de Souza Lima, São Paulo, Brazil
| | - Patricia S Rosa
- Division of Research and Education, Instituto Lauro de Souza Lima, São Paulo, Brazil
| | - Cássio C Ghidella
- Ambulatory of Leprosy, Jardim Guanabara Health Center, Rondonópolis, Brazil
| | - Rodrigo F Ramalho
- Laboratory of Genomics and Molecular Biology, CIPE, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Mabel G Pinilla
- Laboratory of Genomics and Molecular Biology, CIPE, A.C. Camargo Cancer Center, São Paulo, Brazil.,Department of Medical Technology, School of Medicine, University of Concepción, Concepción, Chile
| | - Alex F Carvalho
- Laboratory of Genomics and Molecular Biology, CIPE, A.C. Camargo Cancer Center, São Paulo, Brazil.,Department of Preventive Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Dirce N Carrara
- Laboratory of Genomics and Molecular Biology, CIPE, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Fernando A Soares
- Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Andrea F F Belone
- Department of Anatomic Pathology, Instituto Lauro de Souza Lima, São Paulo, Brazil
| |
Collapse
|
20
|
Ning Z, Zhang K, Zhao L, Lu Y, Sun H, Chen C, Nie X, Lu M, Wang N, Lu Y. Exacerbation of liver steatosis following exposure to famine and overnutrition. Mol Nutr Food Res 2017; 61. [PMID: 28499082 DOI: 10.1002/mnfr.201700097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/13/2017] [Accepted: 05/04/2017] [Indexed: 12/14/2022]
Abstract
SCOPE People suffering from famine in early life and overnutrition in adulthood may have an increased risk for liver steatosis. We aimed to investigate the effects and mechanisms of early nutrition restriction and overnutrition on de novo lipogenesis in the liver. METHODS AND RESULTS Three-wk-old male rats were food restricted for 4 wk and refed a high-fat or normal fat diet individually in metabolic cages for 9 wk. Weight-matched groups were also set up. Fatty acid synthetase expression was measured to estimate de novo lipogenesis in the liver. Parameters of glucose and lipid metabolism were measured with isotope assays. All four groups had comparable body weights. However, the famine high-fat diet group had the highest degree of liver steatosis, the greatest body fat ratio, and insulin resistance. Lipid accumulation, fatty acid synthetase expression, and gluconeogenesis in the liver were significantly higher in the famine and high-fat diet groups (p < 0.05). Moreover, these groups also had markedly lower muscle glucose uptake. CONCLUSION Under famine and high-fat refeeding stress, rats were extremely susceptible to developing hepatic steatosis. This is presumably a consequence of upregulation of de novo lipogenesis and enhanced glucose flux from muscle to de novo lipogenesis in the liver.
Collapse
Affiliation(s)
- Zhiyuan Ning
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Li Zhao
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - You Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Honglin Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaomin Nie
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Meng Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Linoleic acid, α-linolenic acid and enterolactone affect lipid oxidation and expression of lipid metabolism and antioxidant-related genes in hepatic tissue of dairy cows. Br J Nutr 2017. [DOI: 10.1017/s0007114517000976] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
AbstractAlthough beneficial effects have been attributed to PUFA supplementation in high-yielding dairy cows, diets rich in PUFA may also increase oxidative stress in tissues such as the liver. To fully exploit the health benefits of PUFA, we believe that the addition of natural antioxidants could help in preventing oxidative damage. Using an in vitro precision-cut liver slices (PCLS) tissue culture system, we investigated the effects of different linoleic acid (LA, n-6):α-linolenic acid (ALA, n-3) ratios (LA:ALA ratio of 4, LA:ALA ratio of 15 and LA:ALA ratio of 25) in the presence or absence of the antioxidant enterolactone (ENL) on (1) the mRNA abundance of genes with key roles in hepatic lipid metabolism, oxidative stress response and inflammatory processes, (2) oxidative damages to lipids and proteins and (3) superoxide dismutase activity in early-lactating dairy cows. The addition of LA and ALA to PCLS culture media increased oxidative damage to lipids as suggested by higher concentrations of thiobarbituric acid reactive substances and increased the expression of nuclear factor erythroid 2-related factor 2 target genes. The addition of ENL was effective in preventing lipid peroxidation caused by LA and ALA. Transcript abundance of sterol regulatory element-binding transcription factor 1 and its lipogenic target genes acetyl-CoA carboxylase α, fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD) was decreased with LA and ALA, whereas ENL decreased FASN and SCD gene expression. Our results show that addition of LA and ALA to PCLS culture media lowers hepatic lipogenic gene expression and increases oxidative damages to lipids. On the other hand, addition of ENL prevents oxidative damages provoked by these PUFA.
Collapse
|
22
|
Damiano F, Rochira A, Gnoni A, Siculella L. Action of Thyroid Hormones, T3 and T2, on Hepatic Fatty Acids: Differences in Metabolic Effects and Molecular Mechanisms. Int J Mol Sci 2017; 18:ijms18040744. [PMID: 28362337 PMCID: PMC5412329 DOI: 10.3390/ijms18040744] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 12/28/2022] Open
Abstract
The thyroid hormones (THs) 3,3′,5,5′-tetraiodo-l-thyronine (T4) and 3,5,3′-triiodo-l-thyronine (T3) influence many metabolic pathways. The major physiological function of THs is to sustain basal energy expenditure, by acting primarily on carbohydrate and lipid catabolism. Beyond the mobilization and degradation of lipids, at the hepatic level THs stimulate the de novo fatty acid synthesis (de novo lipogenesis, DNL), through both the modulation of gene expression and the rapid activation of cell signalling pathways. 3,5-Diiodo-l-thyronine (T2), previously considered only a T3 catabolite, has been shown to mimic some of T3 effects on lipid catabolism. However, T2 action is more rapid than that of T3, and seems to be independent of protein synthesis. An inhibitory effect on DNL has been documented for T2. Here, we give an overview of the mechanisms of THs action on liver fatty acid metabolism, focusing on the different effects exerted by T2 and T3 on the regulation of the DNL. The inhibitory action on DNL exerted by T2 makes this compound a potential and attractive drug for the treatment of some metabolic diseases and cancer.
Collapse
Affiliation(s)
- Fabrizio Damiano
- Laboratory of Biochemistry and Molecular Biology, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Alessio Rochira
- Laboratory of Biochemistry and Molecular Biology, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Antonio Gnoni
- Department of Basic Medical Sciences, Section of Medical Biochemistry, University of Bari Aldo Moro, 70125 Bari, Italy.
| | - Luisa Siculella
- Laboratory of Biochemistry and Molecular Biology, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| |
Collapse
|
23
|
Chng WBA, Hietakangas V, Lemaitre B. Physiological Adaptations to Sugar Intake: New Paradigms from Drosophila melanogaster. Trends Endocrinol Metab 2017; 28:131-142. [PMID: 27923532 DOI: 10.1016/j.tem.2016.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/30/2016] [Accepted: 11/07/2016] [Indexed: 11/20/2022]
Abstract
Sugars are important energy sources, but high sugar intake poses a metabolic challenge and leads to diseases. Drosophila melanogaster is a generalist fruit breeder that encounters high levels of dietary sugars in its natural habitat. Consequently, Drosophila displays adaptive responses to dietary sugars, including highly conserved and unique metabolic adaptations not described in mammals. Carbohydrate homeostasis is maintained by a network comprising intracellular energy sensors, transcriptional regulators, and hormonal and neuronal mechanisms that together coordinate animal behavior, gut function, and metabolic flux. Here we give an overview of the physiological responses associated with sugar intake and discuss some of the emerging themes and applications of the Drosophila model in understanding sugar sensing and carbohydrate metabolism.
Collapse
Affiliation(s)
- Wen-Bin Alfred Chng
- Global Health Institute, School of Life Sciences, EPFL, Station 19, 1015 Lausanne, Switzerland.
| | - Ville Hietakangas
- Department of Biosciences, University of Helsinki, 00790 Helsinki, Finland; Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, EPFL, Station 19, 1015 Lausanne, Switzerland.
| |
Collapse
|
24
|
Luque GM, Lopez-Vicchi F, Ornstein AM, Brie B, De Winne C, Fiore E, Perez-Millan MI, Mazzolini G, Rubinstein M, Becu-Villalobos D. Chronic hyperprolactinemia evoked by disruption of lactotrope dopamine D2 receptors impacts on liver and adipocyte genes related to glucose and insulin balance. Am J Physiol Endocrinol Metab 2016; 311:E974-E988. [PMID: 27802964 DOI: 10.1152/ajpendo.00200.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 12/21/2022]
Abstract
We studied the impact of high prolactin titers on liver and adipocyte gene expression related to glucose and insulin homeostasis in correlation with obesity onset. To that end we used mutant female mice that selectively lack dopamine type 2 receptors (D2Rs) from pituitary lactotropes (lacDrd2KO), which have chronic high prolactin levels associated with increased body weight, marked increments in fat depots, adipocyte size, and serum lipids, and a metabolic phenotype that intensifies with age. LacDrd2KO mice of two developmental ages, 5 and 10 mo, were used. In the first time point, obesity and increased body weight are marginal, although mice are hyperprolactinemic, whereas at 10 mo there is marked adiposity with a 136% increase in gonadal fat and a 36% increase in liver weight due to lipid accumulation. LacDrd2KO mice had glucose intolerance, hyperinsulinemia, and impaired insulin response to glucose already in the early stages of obesity, but changes in liver and adipose tissue transcription factors were time and tissue dependent. In chronic hyperprolactinemic mice liver Prlr were upregulated, there was liver steatosis, altered expression of the lipogenic transcription factor Chrebp, and blunted response of Srebp-1c to refeeding at 5 mo of age, whereas no effect was observed in the glycogenesis pathway. On the other hand, in adipose tissue a marked decrease in lipogenic transcription factor expression was observed when morbid obesity was already settled. These adaptive changes underscore the role of prolactin signaling in different tissues to promote energy storage.
Collapse
Affiliation(s)
- Guillermina María Luque
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Felicitas Lopez-Vicchi
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Ana María Ornstein
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Belén Brie
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Catalina De Winne
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Esteban Fiore
- Laboratorio de Terapia Génica, Instituto de Investigaciones en Medicina Traslacional (IIMT-CONICET), Universidad Austral, Buenos Aires, Argentina; and
| | - Maria Inés Perez-Millan
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Guillermo Mazzolini
- Laboratorio de Terapia Génica, Instituto de Investigaciones en Medicina Traslacional (IIMT-CONICET), Universidad Austral, Buenos Aires, Argentina; and
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, CONICET, and Departamento de Fisiología, y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, University of Buenos Aires, Argentina
| | - Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina;
| |
Collapse
|
25
|
Athanason MG, Ratliff WA, Chaput D, MarElia CB, Kuehl MN, Stevens SM, Burkhardt BR. Quantitative proteomic profiling reveals hepatic lipogenesis and liver X receptor activation in the PANDER transgenic model. Mol Cell Endocrinol 2016; 436:41-9. [PMID: 27394190 PMCID: PMC5789791 DOI: 10.1016/j.mce.2016.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/06/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022]
Abstract
PANcreatic-DERived factor (PANDER) is a member of a superfamily of FAM3 proteins modulating glycemic levels by metabolic regulation of the liver and pancreas. The precise PANDER-induced hepatic signaling mechanism is still being elucidated and has been very complex due to the pleiotropic nature of this novel hormone. Our PANDER transgenic (PANTG) mouse displays a selective hepatic insulin resistant (SHIR) phenotype whereby insulin signaling is blunted yet lipogenesis is increased, a phenomena observed in type 2 diabetes. To examine the complex PANDER-induced mechanism of SHIR, we utilized quantitative mass spectrometry-based proteomic analysis using Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) to reveal the global hepatic proteome differences within the PANTG under the metabolic states of fasting, fed and insulin-stimulated conditions. Proteomic analysis identified lipid metabolism as one of the top cellular functions differentially altered in all metabolic states. Differentially expressed proteins within the PANTG having a lipid metabolic role included ACC, ACLY, CD36, CYP7A1, FASN and SCD1. Central to the differentially expressed proteins involved in lipid metabolism was the predicted activation of the liver X receptor (LXR) pathway. Western analysis validated the increased hepatic expression of LXRα along with LXR-directed targets such as FASN and CYP7A1 within the PANTG liver. Furthermore, recombinant PANDER was capable of inducing LXR promoter activity in-vitro as determined by luciferase reporter assays. Taken together, PANDER strongly impacts hepatic lipid metabolism across metabolic states and may induce a SHIR phenotype via the LXR pathway.
Collapse
Affiliation(s)
- Mark G Athanason
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Whitney A Ratliff
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Catherine B MarElia
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Melanie N Kuehl
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Stanley M Stevens
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA
| | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, BSF 206, Tampa, FL 33620, USA.
| |
Collapse
|
26
|
Esposito T, Lobaccaro JM, Esposito MG, Monda V, Messina A, Paolisso G, Varriale B, Monda M, Messina G. Effects of low-carbohydrate diet therapy in overweight subjects with autoimmune thyroiditis: possible synergism with ChREBP. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2939-2946. [PMID: 27695291 PMCID: PMC5028075 DOI: 10.2147/dddt.s106440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The thyroid is one of the metabolism regulating glands. Its function is to determine the amount of calories that the body has to burn to maintain normal weight. Thyroiditides are inflammatory processes that mainly result in autoimmune diseases. We have conducted the present study in order to have a clear picture of both autoimmune status and the control of body weight. We have evaluated the amount of either thyroid hormones, or antithyroid, or anti-microsomal, or anti-peroxidase antibodies (Abs) in patients with high amounts of Abs. In a diet devoid of carbohydrates (bread, pasta, fruit, and rice), free from goitrogenic food, and based on body mass index, the distribution of body mass and intracellular and extracellular water conducted for 3 weeks gives the following results: patients treated as above showed a significant reduction of antithyroid (−40%, P<0.013), anti-microsomal (−57%, P<0.003), and anti-peroxidase (−44%, P<0,029) Abs. Untreated patients had a significant increase in antithyroid (+9%, P<0.017) and anti-microsomal (+30%, P<0.028) Abs. Even the level of anti-peroxidase Abs increased without reaching statistical significance (+16%, P>0064). With regard to the body parameters measured in patients who followed this diet, reduction in body weight (−5%, P<0.000) and body mass index (−4%, P<0.000) were observed. Since 83% of patients with high levels of autoantibodies are breath test positive to lactase with a lactase deficit higher than 50%, this fact led us to hypothesize a correlation with carbohydrate-responsive element-binding protein and therefore a possible role of carbohydrate metabolism in the development and maintenance of autoimmune thyroiditis associated with body weight increase and slower basic metabolism.
Collapse
Affiliation(s)
- Teresa Esposito
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine; Laboratory of Molecular Biology and Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Jean Marc Lobaccaro
- UMR, Clermont Université, Centre de Recherche en Nutrition Humaine d'Auvergne, Aubière Cedex, France
| | | | - Vincenzo Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine
| | - Antonietta Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine
| | - Giuseppe Paolisso
- Department of Scienze Mediche, Chirurgiche, Neurologiche, Metaboliche e dell'Invecchiamento, Second University of Naples, Naples
| | - Bruno Varriale
- Laboratory of Molecular Biology and Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine
| | - Giovanni Messina
- Department of Experimental Medicine, Section of Human Physiology and Unit of Dietetics and Sports Medicine; Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
27
|
Logan SM, Storey KB. Tissue-specific response of carbohydrate-responsive element binding protein (ChREBP) to mammalian hibernation in 13-lined ground squirrels. Cryobiology 2016; 73:103-11. [PMID: 27614289 DOI: 10.1016/j.cryobiol.2016.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 08/31/2016] [Accepted: 09/06/2016] [Indexed: 11/18/2022]
Abstract
Mammalian hibernation is characterized by a general suppression of energy expensive processes and a switch to lipid oxidation as the primary fuel source. Glucose-responsive carbohydrate responsive element binding protein (ChREBP) has yet to be studied in hibernating organisms, which prepare for the cold winter months by feeding until they exhibit an obesity-like state that is accompanied by naturally-induced and completely reversible insulin resistance. Studying ChREBP expression and activity in the hibernating 13-lined ground squirrel is important to better understand the molecular mechanisms that regulate energy metabolism under cellular stress. Immunoblotting was used to determine the relative expression level and subcellular localization of ChREBP, as well as serine phosphorylation at 95 kDa, comparing euthermic and late torpid ground squirrel liver, kidney, heart and muscle. DNA-binding ELISAs and RT-PCR were used to explore ChREBP transcriptional activity during cold stress. ChREBP activity seemed generally suppressed in liver and kidney. During torpor, ChREBP total protein levels decreased to 44% of EC in liver, phosphoserine levels increased 2.1-fold of EC in kidney, and downstream Fasn/Pkl transcript levels decreased to <60% of EC in liver. By contrast, ChREBP activity generally increased during torpor in cardiac and skeletal muscle, where ChREBP total protein levels increased over 1.5-fold and 5-fold of EC in muscle and heart, respectively; where DNA-binding increased by ∼2-fold of EC in muscle; and where Fasn transcript levels increased over 3-fold and 7-fold in both muscle and heart, respectively. In summary, ChREBP has a tissue-specific role in regulating energy metabolism during hibernation.
Collapse
Affiliation(s)
- Samantha M Logan
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Kenneth B Storey
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| |
Collapse
|
28
|
Adipose tissue mTORC2 regulates ChREBP-driven de novo lipogenesis and hepatic glucose metabolism. Nat Commun 2016; 7:11365. [PMID: 27098609 PMCID: PMC4844681 DOI: 10.1038/ncomms11365] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/18/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue de novo lipogenesis (DNL) positively influences insulin sensitivity, is reduced in obesity, and predicts insulin resistance. Therefore, elucidating mechanisms controlling adipose tissue DNL could lead to therapies for type 2 diabetes. Here, we report that mechanistic target of rapamycin complex 2 (mTORC2) functions in white adipose tissue (WAT) to control expression of the lipogenic transcription factor ChREBPβ. Conditionally deleting the essential mTORC2 subunit Rictor in mature adipocytes decreases ChREBPβ expression, which reduces DNL in WAT, and impairs hepatic insulin sensitivity. Mechanistically, Rictor/mTORC2 promotes ChREBPβ expression in part by controlling glucose uptake, but without impairing pan-AKT signalling. High-fat diet also rapidly decreases adipose tissue ChREBPβ expression and insulin sensitivity in wild-type mice, and does not further exacerbate insulin resistance in adipose tissue Rictor knockout mice, implicating adipose tissue DNL as an early target in diet-induced insulin resistance. These data suggest mTORC2 functions in WAT as part of an extra-hepatic nutrient-sensing mechanism to control glucose homeostasis. The kinase mTOR controls anabolic metabolism. Here, the authors create fat-specific mTORC2 knockout mice using the Adiponectin-Cre driver and show mTORC2 signalling is important for systemic metabolic homeostasis by controlling adipocyte de novo lipogenesis and glucose uptake.
Collapse
|
29
|
Singh P, Irwin DM. Contrasting Patterns in the Evolution of Vertebrate MLX Interacting Protein (MLXIP) and MLX Interacting Protein-Like (MLXIPL) Genes. PLoS One 2016; 11:e0149682. [PMID: 26910886 PMCID: PMC4766361 DOI: 10.1371/journal.pone.0149682] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/03/2016] [Indexed: 01/09/2023] Open
Abstract
ChREBP and MondoA are glucose-sensitive transcription factors that regulate aspects of energy metabolism. Here we performed a phylogenomic analysis of Mlxip (encoding MondoA) and Mlxipl (encoding ChREBP) genes across vertebrates. Analysis of extant Mlxip and Mlxipl genes suggests that the most recent common ancestor of these genes was composed of 17 coding exons. Single copy genes encoding both ChREBP and MondoA, along with their interacting partner Mlx, were found in diverse vertebrate genomes, including fish that have experienced a genome duplication. This observation suggests that a single Mlx gene has been retained to maintain coordinate regulation of ChREBP and MondoA. The ChREBP-β isoform, the more potent and constitutively active isoform, appeared with the evolution of tetrapods and is absent from the Mlxipl genes of fish. Evaluation of the conservation of ChREBP and MondoA sequences demonstrate that MondoA is better conserved and potentially mediates more ancient function in glucose metabolism.
Collapse
Affiliation(s)
- Parmveer Singh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
30
|
Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis 2016; 247:225-82. [PMID: 26967715 DOI: 10.1016/j.atherosclerosis.2016.02.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 02/02/2016] [Indexed: 02/08/2023]
Abstract
The growing worldwide prevalence of overnutrition and underexertion threatens the gains that we have made against atherosclerotic cardiovascular disease and other maladies. Chronic overnutrition causes the atherometabolic syndrome, which is a cluster of seemingly unrelated health problems characterized by increased abdominal girth and body-mass index, high fasting and postprandial concentrations of cholesterol- and triglyceride-rich apoB-lipoproteins (C-TRLs), low plasma HDL levels, impaired regulation of plasma glucose concentrations, hypertension, and a significant risk of developing overt type 2 diabetes mellitus (T2DM). In addition, individuals with this syndrome exhibit fatty liver, hypercoagulability, sympathetic overactivity, a gradually rising set-point for body adiposity, a substantially increased risk of atherosclerotic cardiovascular morbidity and mortality, and--crucially--hyperinsulinemia. Many lines of evidence indicate that each component of the atherometabolic syndrome arises, or is worsened by, pathway-selective insulin resistance and responsiveness (SEIRR). Individuals with SEIRR require compensatory hyperinsulinemia to control plasma glucose levels. The result is overdrive of those pathways that remain insulin-responsive, particularly ERK activation and hepatic de-novo lipogenesis (DNL), while carbohydrate regulation deteriorates. The effects are easily summarized: if hyperinsulinemia does something bad in a tissue or organ, that effect remains responsive in the atherometabolic syndrome and T2DM; and if hyperinsulinemia might do something good, that effect becomes resistant. It is a deadly imbalance in insulin action. From the standpoint of human health, it is the worst possible combination of effects. In this review, we discuss the origins of the atherometabolic syndrome in our historically unprecedented environment that only recently has become full of poorly satiating calories and incessant enticements to sit. Data are examined that indicate the magnitude of daily caloric imbalance that causes obesity. We also cover key aspects of healthy, balanced insulin action in liver, endothelium, brain, and elsewhere. Recent insights into the molecular basis and pathophysiologic harm from SEIRR in these organs are discussed. Importantly, a newly discovered oxide transport chain functions as the master regulator of the balance amongst different limbs of the insulin signaling cascade. This oxide transport chain--abbreviated 'NSAPP' after its five major proteins--fails to function properly during chronic overnutrition, resulting in this harmful pattern of SEIRR. We also review the origins of widespread, chronic overnutrition. Despite its apparent complexity, one factor stands out. A sophisticated junk food industry, aided by subsidies from willing governments, has devoted years of careful effort to promote overeating through the creation of a new class of food and drink that is low- or no-cost to the consumer, convenient, savory, calorically dense, yet weakly satiating. It is past time for the rest of us to overcome these foes of good health and solve this man-made epidemic.
Collapse
|
31
|
Santamarina AB, Oliveira JL, Silva FP, Carnier J, Mennitti LV, Santana AA, de Souza GHI, Ribeiro EB, Oller do Nascimento CM, Lira FS, Oyama LM. Green Tea Extract Rich in Epigallocatechin-3-Gallate Prevents Fatty Liver by AMPK Activation via LKB1 in Mice Fed a High-Fat Diet. PLoS One 2015; 10:e0141227. [PMID: 26536464 PMCID: PMC4633218 DOI: 10.1371/journal.pone.0141227] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 10/05/2015] [Indexed: 01/19/2023] Open
Abstract
Supplementation with epigallocatechin-3-gallate has been determined to aid in the prevention of obesity. Decaffeinated green tea extract appears to restore a normal hepatic metabolic profile and attenuate high-fat diet (HFD)-induced effects, thereby preventing non-alcoholic fatty liver disease in mice. Mice were maintained on either a control diet (CD) or HFD for 16 weeks and supplemented with either water or green tea extract (50 mg/kg/day). The body mass increase, serum adiponectin level, and lipid profile were measured over the course of the treatment. Furthermore, the AMPK pathway protein expression in the liver was measured. From the fourth week, the weight gain in the CD + green tea extract (CE) group was lower than that in the CD + water (CW) group. From the eighth week, the weight gain in the HFD + water (HFW) group was found to be higher than that in the CW group. Moreover, the weight gain in the HFD + green tea extract (HFE) group was found to be lower than that in the HFW group. Carcass lipid content was found to be higher in the HFW group than that in the CW and HFE groups. Serum analysis showed reduced non-esterified fatty acid level in the CE and HFE groups as compared with their corresponding placebo groups. Increased adiponectin level was observed in the same groups. Increased VLDL-TG secretion was observed in the HFW group as compared with the CW and HFE groups. Increased protein expression of AdipoR2, SIRT1, pLKB1, and pAMPK was observed in the HFE group, which explained the reduced expression of ACC, FAS, SREBP-1, and ChREBP in this group. These results indicate that the effects of decaffeinated green tea extract may be related to the activation of AMPK via LKB1 in the liver of HFD-fed mice.
Collapse
Affiliation(s)
| | - Juliana L. Oliveira
- Departamento de Fisiologia—Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda P. Silva
- Departamento de Fisiologia—Universidade Federal de São Paulo, São Paulo, Brazil
| | - June Carnier
- Departamento de Fisiologia—Universidade Federal de São Paulo, São Paulo, Brazil
| | - Laís V. Mennitti
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde—Universidade Federal de São Paulo, Santos, Brazil
| | - Aline A. Santana
- Departamento de Fisiologia—Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Eliane B. Ribeiro
- Departamento de Fisiologia—Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Fábio S. Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Universidade Estadual Paulista, UNESP, Presidente Prudente, SP, Brazil
| | - Lila M. Oyama
- Departamento de Fisiologia—Universidade Federal de São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
32
|
Ratliff WA, Athanason MG, Chechele AC, Kuehl MN, Fernandez AM, MarElia CB, Burkhardt BR. Hepatic nutrient and hormonal regulation of the PANcreatic-DERived factor (PANDER) promoter. Mol Cell Endocrinol 2015; 413:101-12. [PMID: 26123584 DOI: 10.1016/j.mce.2015.05.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 04/29/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
PANcreatic-DERived factor (PANDER, FAM3B) has been shown to regulate glycemic levels via interactions with both pancreatic islets and the liver. Although PANDER is predominantly expressed from the endocrine pancreas, recent work has provided sufficient evidence that the liver may also be an additional tissue source of PANDER production. At physiological levels, PANDER is capable of disrupting insulin signaling and promoting increased hepatic glucose production. As shown in some animal models, strong expression of PANDER, induced by viral delivery within the liver, induces hepatic steatosis. However, no studies to date have explicitly characterized the transcriptional regulation of PANDER from the liver. Therefore, our investigation elucidated the nutrient and hormonal regulation of the hepatic PANDER promoter. Initial RNA-ligated rapid amplification of cDNA ends identified a novel transcription start site (TSS) approximately 26 bp upstream of the PANDER translational start codon not previously revealed in pancreatic β-cell lines. Western evaluation of various murine tissues demonstrated robust expression in the liver and brain. Promoter analysis identified strong tissue-specific activity of the PANDER promoter in both human and murine liver-derived cell lines. The minimal element responsible for maximal promoter activity within hepatic cell lines was located between -293 and -3 of the identified TSS. PANDER promoter activity was inhibited by both insulin and palmitate, whereas glucose strongly increased expression. The minimal element was responsible for maximal glucose-responsive and basal activity. Co-transfection reporter assays, chromatin-immunoprecipitation (ChIP) and site-directed mutagenesis revealed that the carbohydrate-responsive element binding protein (ChREBP) increased PANDER promoter activity and interacted with the PANDER promoter. E-box 3 was shown to be critical for basal and glucose responsive expression. In summary, in-vitro and in-vivo glucose is a potent stimulator of the PANDER promoter within the liver and this response may be facilitated by ChREBP.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Mark G Athanason
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Alicia C Chechele
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Melanie N Kuehl
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Amanda M Fernandez
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Catherine B MarElia
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA.
| |
Collapse
|
33
|
Turtle anoxia tolerance: Biochemistry and gene regulation. Biochim Biophys Acta Gen Subj 2015; 1850:1188-96. [DOI: 10.1016/j.bbagen.2015.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/01/2015] [Indexed: 12/16/2022]
|
34
|
Wu W, Tsuchida H, Kato T, Niwa H, Horikawa Y, Takeda J, Iizuka K. Fat and carbohydrate in western diet contribute differently to hepatic lipid accumulation. Biochem Biophys Res Commun 2015; 461:681-6. [PMID: 25931000 DOI: 10.1016/j.bbrc.2015.04.092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 04/19/2015] [Indexed: 01/16/2023]
Abstract
We investigated the contributions of dietary fat and dietary carbohydrate to the development of fatty liver induced by western diet (WD). Compared with WD-fed wild type (WT) mice, livers of WD-fed ChREBP(-/-) mice showed lipid droplets of varying sizes around the hepatic lobules, while hepatic triglyceride and cholesterol contents were only modestly decreased. Inflammation and fibrosis were suppressed in ChREBP(-/-) mice. In addition, compared with WD-fed WT mice, ChREBP(-/-) mice showed decreased β-oxidation, ketogenesis and FGF21 production, increased intestinal lipid absorption, and decreased VLDL secretion. These findings suggest that dietary fat and carbohydrate contribute differently to the development of fatty liver.
Collapse
Affiliation(s)
- Wudelehu Wu
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Hiromi Tsuchida
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Takehiro Kato
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; Matsunami General Hospital, Gifu 501-6062, Japan
| | - Horoyuki Niwa
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Yukio Horikawa
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Jun Takeda
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | - Katsumi Iizuka
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; Gifu University Hospital Center for Nutritional Support and Infection Control, Gifu 501-1194, Japan.
| |
Collapse
|
35
|
Tsai CY, Peh MT, Feng W, Dymock BW, Moore PK. Hydrogen sulfide promotes adipogenesis in 3T3L1 cells. PLoS One 2015; 10:e0119511. [PMID: 25822632 PMCID: PMC4378953 DOI: 10.1371/journal.pone.0119511] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 01/14/2015] [Indexed: 12/30/2022] Open
Abstract
The effect of hydrogen sulfide (H2S) on differentiation of 3T3L1-derived adipocytes was examined. Endogenous H2S was increased after 3T3L1 differentiation. The expression of the H2S-synthesising enzymes, cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST), was increased in a time-dependent manner during 3T3L1 differentiation. Expression of genes associated with adipogenesis related genes including fatty acid binding protein 4 (FABP4/aP2), a key regulator of this process, was increased by GYY4137 (a slow-releasing H2S donor compound) and sodium hydrosulfide (NaHS, a classical H2S donor) but not by ZYJ1122 or time-expired NaHS. Furthermore expression of these genes were reduced by aminooxyacetic acid (AOAA, CBS inhibitor), DL-propargylglycine (PAG, CSE inhibitor) as well as by CSE small interference RNA (siCSE) and siCBS. The size and number of lipid droplets in mature adipocytes was significantly increased by both GYY4137 and NaHS, which also impaired the ability of CL316,243 (β3-agonist) to promote lipolysis in these cells. In contrast, AOAA and PAG had the opposite effect. Taken together, we show that the H2S-synthesising enzymes CBS, CSE and 3-MST are endogenously expressed during adipogenesis and that both endogenous and exogenous H2S modulate adipogenesis and adipocyte maturation.
Collapse
Affiliation(s)
- Chin-Yi Tsai
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
- * E-mail:
| | - Meng Teng Peh
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Wei Feng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Brian William Dymock
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Philip Keith Moore
- Neurobiology Group, Life Science Institute, Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, Singapore
| |
Collapse
|
36
|
Zhang C, Cooper DE, Grevengoed TJ, Li LO, Klett EL, Eaton JM, Harris TE, Coleman RA. Glycerol-3-phosphate acyltransferase-4-deficient mice are protected from diet-induced insulin resistance by the enhanced association of mTOR and rictor. Am J Physiol Endocrinol Metab 2014; 307:E305-15. [PMID: 24939733 PMCID: PMC4121579 DOI: 10.1152/ajpendo.00034.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Glycerol-3-phosphate acyltransferase (GPAT) activity is highly induced in obese individuals with insulin resistance, suggesting a correlation between GPAT function, triacylglycerol accumulation, and insulin resistance. We asked whether microsomal GPAT4, an isoform regulated by insulin, might contribute to the development of hepatic insulin resistance. Compared with control mice fed a high fat diet, Gpat4(-/-) mice were more glucose tolerant and were protected from insulin resistance. Overexpression of GPAT4 in mouse hepatocytes impaired insulin-suppressed gluconeogenesis and insulin-stimulated glycogen synthesis. Impaired glucose homeostasis was coupled to inhibited insulin-stimulated phosphorylation of Akt(Ser⁴⁷³) and Akt(Thr³⁰⁸). GPAT4 overexpression inhibited rictor's association with the mammalian target of rapamycin (mTOR), and mTOR complex 2 (mTORC2) activity. Compared with overexpressed GPAT3 in mouse hepatocytes, GPAT4 overexpression increased phosphatidic acid (PA), especially di16:0-PA. Conversely, in Gpat4(-/-) hepatocytes, both mTOR/rictor association and mTORC2 activity increased, and the content of PA in Gpat4(-/-) hepatocytes was lower than in controls, with the greatest decrease in 16:0-PA species. Compared with controls, liver and skeletal muscle from Gpat4(-/-)-deficient mice fed a high-fat diet were more insulin sensitive and had a lower hepatic content of di16:0-PA. Taken together, these data demonstrate that a GPAT4-derived lipid signal, likely di16:0-PA, impairs insulin signaling in mouse liver and contributes to hepatic insulin resistance.
Collapse
Affiliation(s)
- Chongben Zhang
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Daniel E Cooper
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Trisha J Grevengoed
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Lei O Li
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Eric L Klett
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | - James M Eaton
- Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia
| | - Rosalind A Coleman
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina;
| |
Collapse
|
37
|
Aguilar-Salinas CA, Tusie-Luna T, Pajukanta P. Genetic and environmental determinants of the susceptibility of Amerindian derived populations for having hypertriglyceridemia. Metabolism 2014; 63:887-94. [PMID: 24768220 PMCID: PMC4315146 DOI: 10.1016/j.metabol.2014.03.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/22/2014] [Accepted: 03/25/2014] [Indexed: 12/31/2022]
Abstract
Here, we discuss potential explanations for the higher prevalence of hypertriglyceridemia in populations with an Amerindian background. Although environmental factors are the triggers, the search for the ethnic related factors that explain the increased susceptibility of the Amerindians is a promising area for research. The study of the genetics of hypertriglyceridemia in Hispanic populations faces several challenges. Ethnicity could be a major confounding variable to prove genetic associations. Despite that, the study of hypertriglyceridemia in Hispanics has resulted in significant contributions. Two GWAS reports have exclusively included Mexican mestizos. Fifty percent of the associations reported in Caucasians could be generalized to the Mexicans, but in many cases the Mexican lead SNP was different than that reported in Europeans. Both reports included new associations with apo B or triglycerides concentrations. The frequency of susceptibility alleles in Mexicans is higher than that found in Europeans for several of the genes with the greatest effect on triglycerides levels. An example is the SNP rs964184 in APOA5. The same trend was observed for ANGPTL3 and TIMD4 variants. In summary, we postulate that the study of the genetic determinants of hypertriglyceridemia in Amerindian populations which have major changes in their lifestyle, may prove to be a great resource to identify new genes and pathways associated with hypertriglyceridemia.
Collapse
Affiliation(s)
- Carlos A Aguilar-Salinas
- Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición, México City, México.
| | - Teresa Tusie-Luna
- Unit of Molecular Biology and Genomic Medicine, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición, México City, México.
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA; Molecular Biology Institute at UCLA, Los Angeles, USA.
| |
Collapse
|
38
|
Krivoruchko A, Storey KB. Activation of the carbohydrate response element binding protein (ChREBP) in response to anoxia in the turtle Trachemys scripta elegans. Biochim Biophys Acta Gen Subj 2014; 1840:3000-5. [PMID: 24931694 DOI: 10.1016/j.bbagen.2014.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 06/04/2014] [Accepted: 06/05/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND ChREBP (carbohydrate response element binding protein) is a glucose-responsive transcription factor that is known to be an important regulator of glycolytic and lipogenic genes in response to glucose. We hypothesized that activation of ChREBP could be relevant to anoxia survival by the anoxia-tolerant turtle, Trachemys scripta elegans. METHODS Expression of ChREBP in response to 5 and 20h of anoxia was examined using RT-PCR and Western immunoblotting. In addition, subcellular localization and DNA-binding activity of ChREBP protein were assessed and transcript levels of liver pyruvate kinase (LPK), a downstream gene under ChREBP control were quantified using RT-PCR. RESULTS ChREBP was anoxia-responsive in kidney and liver, with transcript levels increasing by 1.2-1.8 fold in response to anoxia and protein levels increasing by 1.8-1.9 fold. Enhanced nuclear presence under anoxia was also observed in both tissues by 2.2-2.8 fold. A 4.2 fold increase in DNA binding activity of ChREBP was also observed in liver in response to 5h of anoxia. In addition, transcript levels of LPK increased by 2.1 fold in response to 5h of anoxia in the liver. CONCLUSIONS The results suggest that activation of ChREBP in response to anoxia might be a crucial factor for anoxia survival in turtle liver by contributing to elevated glycolytic flux in the initial phases of oxygen limitation. GENERAL SIGNIFICANCE This study provides the first demonstration of activation of ChREBP in response to anoxia in a natural model of anoxia tolerance, further improving our understanding of the molecular nature of anoxia tolerance.
Collapse
Affiliation(s)
- Anastasia Krivoruchko
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada.
| | - Kenneth B Storey
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| |
Collapse
|
39
|
Wang XL, Wen XF, Li RB, Liu B, Qiu GM, Wen JL, Wang YM. Chrebp regulates the transcriptional activity of androgen receptor in prostate cancer. Tumour Biol 2014; 35:8143-8. [PMID: 24845031 DOI: 10.1007/s13277-014-2085-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/10/2014] [Indexed: 01/27/2023] Open
Abstract
Androgen receptor (AR), a member of nuclear hormone receptor, plays an essential role in the initiation and progression of prostate cancer (PCa). In the present study, by way of immunoprecipitation followed by mass spectrometry (IP/MS) system, we found that carbohydrate-responsive element-binding protein (Chrebp), a glucose sensor in normal and cancer cells, interacted with AR in LNCaP cells. The interaction was further confirmed by coimmunoprecipitation analysis. Besides, Chrebp is required for the optimal transcriptional activity of AR in promoting the transcription of the prostate-specific antigen (PSA) promoter and messenger RNA (mRNA) expression. Consistently, knockdown of Chrebp using small interfering RNA (siRNA) in LNCaP cells reduced endogenous PSA levels. Together, our study demonstrates that Chrebp interacts with AR and regulates its transcriptional activity.
Collapse
Affiliation(s)
- Xue-Lei Wang
- Department of Urology, East Hospital, Tongji University School of Medicine, Jimo Road 150, 200120, Shanghai, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Karagianni P, Talianidis I. Transcription factor networks regulating hepatic fatty acid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1851:2-8. [PMID: 24814048 DOI: 10.1016/j.bbalip.2014.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 02/06/2023]
Abstract
Tight regulation of lipid levels is critical for cellular and organismal homeostasis, not only in terms of energy utilization and storage, but also to prevent potential toxicity. The liver utilizes a set of hepatic transcription factors to regulate the expression of genes implicated in all aspects of lipid metabolism including catabolism, transport, and synthesis. In this article, we will review the main transcriptional mechanisms regulating the expression of genes involved in hepatic lipid metabolism. The principal regulatory pathways are composed of simple modules of transcription factor crosstalks, which correspond to building blocks of more complex regulatory networks. These transcriptional networks contribute to the regulation of proper lipid homeostasis in parallel to posttranslational mechanisms and end product-mediated modulation of lipid metabolizing enzymes. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.
Collapse
Affiliation(s)
| | - Iannis Talianidis
- Biomedical Sciences Research Center Alexander Fleming, 16672 Vari, Greece.
| |
Collapse
|
41
|
Link JM, Hurlin PJ. The activities of MYC, MNT and the MAX-interactome in lymphocyte proliferation and oncogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:554-62. [PMID: 24731854 DOI: 10.1016/j.bbagrm.2014.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/25/2014] [Accepted: 04/04/2014] [Indexed: 12/29/2022]
Abstract
The MYC family of proteins plays essential roles in embryonic development and in oncogenesis. Efforts over the past 30 years to define the transcriptional activities of MYC and how MYC functions to promote proliferation have produced evolving models of MYC function. One picture that has emerged of MYC and its partner protein MAX is of a transcription factor complex with a seemingly unique ability to stimulate the transcription of genes that are epigenetically poised for transcription and to amplify the transcription of actively transcribed genes. During lymphocyte activation, MYC is upregulated and stimulates a pro-proliferative program in part through the upregulation of a wide variety of metabolic effector genes that facilitate cell growth and cell cycle progression. MYC upregulation simultaneously sensitizes cells to apoptosis and activated lymphocytes and lymphoma cells have pro-survival attributes that allow MYC-driven proliferation to prevail. For example, the MAX-interacting protein MNT is upregulated in activated lymphocytes and was found to protect lymphocytes from MYC-dependent apoptosis. Here we review the activities of MYC, MNT and other MAX interacting proteins in the setting of T and B cell activation and oncogenesis. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.
Collapse
Affiliation(s)
- Jason M Link
- Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Peter J Hurlin
- Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA; Department of Cell and Developmental Biology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
42
|
Corella D, Ordovás JM. How does the Mediterranean diet promote cardiovascular health? Current progress toward molecular mechanisms: gene-diet interactions at the genomic, transcriptomic, and epigenomic levels provide novel insights into new mechanisms. Bioessays 2014; 36:526-37. [PMID: 24706458 DOI: 10.1002/bies.201300180] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epidemiological evidence supports a health-promoting effect of the Mediterranean Diet (MedDiet), especially in the prevention of cardiovascular diseases. These cardiovascular benefits have been attributed to a number of components of the MedDiet such as monounsaturated fatty acids, antioxidant vitamins and phytochemicals. However, the underlying mechanisms remain unknown. Likewise, little is known about the genes that define inter-individual variation in response to the MedDiet, although the TCF7L2 gene is emerging as an illustrative candidate for determining relative risk of cardiovascular events in response to the MedDiet. Moreover, omics technologies are providing evidence supporting potential mechanisms, some of them implicating epigenetics (i.e. microRNAs, methylation), and certain data suggest that some traditional foods could contribute via microRNAs possibly acting as exogenous regulators of gene expression. Future research should aim at increasing and consolidating the nutrigenetic and nutrigenomic knowledge of the MedDiet in order to provide sound, personalized and optimized nutritional recommendations.
Collapse
Affiliation(s)
- Dolores Corella
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
43
|
Johnson DW, Llop JR, Farrell SF, Yuan J, Stolzenburg LR, Samuelson AV. The Caenorhabditis elegans Myc-Mondo/Mad complexes integrate diverse longevity signals. PLoS Genet 2014; 10:e1004278. [PMID: 24699255 PMCID: PMC3974684 DOI: 10.1371/journal.pgen.1004278] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 02/18/2014] [Indexed: 11/29/2022] Open
Abstract
The Myc family of transcription factors regulates a variety of biological processes, including the cell cycle, growth, proliferation, metabolism, and apoptosis. In Caenorhabditis elegans, the "Myc interaction network" consists of two opposing heterodimeric complexes with antagonistic functions in transcriptional control: the Myc-Mondo:Mlx transcriptional activation complex and the Mad:Max transcriptional repression complex. In C. elegans, Mondo, Mlx, Mad, and Max are encoded by mml-1, mxl-2, mdl-1, and mxl-1, respectively. Here we show a similar antagonistic role for the C. elegans Myc-Mondo and Mad complexes in longevity control. Loss of mml-1 or mxl-2 shortens C. elegans lifespan. In contrast, loss of mdl-1 or mxl-1 increases longevity, dependent upon MML-1:MXL-2. The MML-1:MXL-2 and MDL-1:MXL-1 complexes function in both the insulin signaling and dietary restriction pathways. Furthermore, decreased insulin-like/IGF-1 signaling (ILS) or conditions of dietary restriction increase the accumulation of MML-1, consistent with the notion that the Myc family members function as sensors of metabolic status. Additionally, we find that Myc family members are regulated by distinct mechanisms, which would allow for integrated control of gene expression from diverse signals of metabolic status. We compared putative target genes based on ChIP-sequencing data in the modENCODE project and found significant overlap in genomic DNA binding between the major effectors of ILS (DAF-16/FoxO), DR (PHA-4/FoxA), and Myc family (MDL-1/Mad/Mxd) at common target genes, which suggests that diverse signals of metabolic status converge on overlapping transcriptional programs that influence aging. Consistent with this, there is over-enrichment at these common targets for genes that function in lifespan, stress response, and carbohydrate metabolism. Additionally, we find that Myc family members are also involved in stress response and the maintenance of protein homeostasis. Collectively, these findings indicate that Myc family members integrate diverse signals of metabolic status, to coordinate overlapping metabolic and cytoprotective transcriptional programs that determine the progression of aging.
Collapse
Affiliation(s)
- David W. Johnson
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Jesse R. Llop
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Sara F. Farrell
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Jie Yuan
- Rochester Institute of Technology, Computer Science Department, Rochester, New York, United States of America
| | - Lindsay R. Stolzenburg
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Andrew V. Samuelson
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| |
Collapse
|
44
|
Ritze Y, Bárdos G, Claus A, Ehrmann V, Bergheim I, Schwiertz A, Bischoff SC. Lactobacillus rhamnosus GG protects against non-alcoholic fatty liver disease in mice. PLoS One 2014; 9:e80169. [PMID: 24475018 PMCID: PMC3903470 DOI: 10.1371/journal.pone.0080169] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 10/08/2013] [Indexed: 11/29/2022] Open
Abstract
Objective Experimental evidence revealed that obesity-associated non-alcoholic fatty liver disease (NAFLD) is linked to changes in intestinal permeability and translocation of bacterial products to the liver. Hitherto, no reliable therapy is available except for weight reduction. Within this study, we examined the possible effect of the probiotic bacterial strain Lactobacillus rhamnosus GG (LGG) as protective agent against experimental NAFLD in a mouse model. Methods Experimental NAFLD was induced by a high-fructose diet over eight weeks in C57BL/J6 mice. Fructose was administered via the drinking water containing 30% fructose with or without LGG at a concentration resulting in approximately 5×107 colony forming units/g body weight. Mice were examined for changes in small intestinal microbiota, gut barrier function, lipopolysaccharide (LPS) concentrations in the portal vein, liver inflammation and fat accumulation in the liver. Results LGG increased beneficial bacteria in the distal small intestine. Moreover, LGG reduced duodenal IκB protein levels and restored the duodenal tight junction protein concentration. Portal LPS (P≤0.05) was reduced and tended to attenuate TNF-α, IL-8R and IL-1β mRNA expression in the liver feeding a high-fructose diet supplemented with LGG. Furthermore liver fat accumulation and portal alanine-aminotransferase concentrations (P≤0.05) were attenuated in mice fed the high-fructose diet and LGG. Conclusions We show for the first time that LGG protects mice from NAFLD induced by a high-fructose diet. The underlying mechanisms of protection likely involve an increase of beneficial bacteria, restoration of gut barrier function and subsequent attenuation of liver inflammation and steatosis.
Collapse
Affiliation(s)
- Yvonne Ritze
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Gyöngyi Bárdos
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Anke Claus
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Veronika Ehrmann
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Ina Bergheim
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
- Department of Nutritional Science, Friedrich-Schiller-University, Jena, Germany
| | | | - Stephan C. Bischoff
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
45
|
Xiao C, Dash S, Morgantini C, Lewis GF. New and emerging regulators of intestinal lipoprotein secretion. Atherosclerosis 2014; 233:608-615. [PMID: 24534456 DOI: 10.1016/j.atherosclerosis.2013.12.047] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/12/2013] [Accepted: 12/31/2013] [Indexed: 12/25/2022]
Abstract
Overproduction of hepatic apoB100-containing VLDL particles has been well documented in animal models and in humans with insulin resistance such as the metabolic syndrome and type 2 diabetes, and contributes to the typical dyslipidemia of these conditions. In addition, postprandial hyperlipidemia and elevated plasma concentrations of intestinal apoB48-containing chylomicron and chylomicron remnant particles have been demonstrated in insulin resistant states. Intestinal lipoprotein production is primarily determined by the amount of fat ingested and absorbed. Until approximately 10 years ago, however, relatively little attention was paid to the role of the intestine itself in regulating the production of triglyceride-rich lipoproteins (TRL) and its dysregulation in pathological states such as insulin resistance. We and others have shown that insulin resistant animal models and humans are characterized by overproduction of intestinal apoB48-containing lipoproteins. Whereas various factors are known to regulate hepatic lipoprotein particle production, less is known about factors that regulate the production of intestinal lipoprotein particles. Monosacharides, plasma free fatty acids (FFA), resveratrol, intestinal peptides (e.g. GLP-1 and GLP-2), and pancreatic hormones (e.g. insulin) have recently been shown to be important regulators of intestinal lipoprotein secretion. Available evidence in humans and animal models strongly supports the concept that the small intestine is not merely an absorptive organ but rather plays an active role in regulating the rate of production of chylomicrons in fed and fasting states. Metabolic signals in insulin resistance and type 2 diabetes and in some cases an aberrant intestinal response to these factors contribute to the enhanced formation and secretion of TRL. Understanding the regulation of intestinal lipoprotein production is imperative for the development of new therapeutic strategies for the prevention and treatment of dyslipidemia. Here we review recent developments in this field and present evidence that intestinal lipoprotein production is a process with metabolic plasticity and that modulation of intestinal lipoprotein secretion may be a feasible therapeutic strategy in the treatment of dyslipidemia and possibly prevention of atherosclerosis.
Collapse
Affiliation(s)
- Changting Xiao
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada
| | - Satya Dash
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada
| | - Cecilia Morgantini
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada
| | - Gary F Lewis
- Department of Medicine, University of Toronto, Canada; Department of Physiology, University of Toronto, Canada; Banting and Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
46
|
Abstract
Obesity is a global health problem that increases the risk of several common diseases. Liver receptor homologue-1 (LRH-1) has an important role in steroid hormone metabolism, which influences body weight. Whether LRH-1 gene deletion causes obesity is yet to be clarified. In this study using LRH-1 heterozygous knockout (LRH-1(+/-)) mice, we investigated the role of LRH-1 on body weight gain and glucose and lipid metabolism. LRH-1(+/-) mice showed mild but significant body weight gains compared with wild-type littermate mice after being fed a high-fat diet. We performed glucose tolerance tests and insulin tolerance tests and did not find any significant differences between wild-type and LRH-1(+/-) mice. To clarify how LRH-1 gene deletion affects body weight gain, we measured food intake, oxygen consumption, respiratory quotient, spontaneous activity and rectal temperature, and found no significant differences between wild-type and LRH-1(+/-) mice fed a normal diet and a high-fat diet. The results suggest that heterozygous gene deletion of LRH-1 causes body weight gains without any apparent worsening of glucose and lipid metabolism. Identifying the effects of LRH-1 on body weight will aid in understanding the pathogenesis of obesity.
Collapse
Affiliation(s)
- Taisuke Hattori
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan
| | | | | | | |
Collapse
|
47
|
Cortés VA, Cautivo KM, Rong S, Garg A, Horton JD, Agarwal AK. Leptin ameliorates insulin resistance and hepatic steatosis in Agpat2-/- lipodystrophic mice independent of hepatocyte leptin receptors. J Lipid Res 2013; 55:276-88. [PMID: 24293639 DOI: 10.1194/jlr.m045799] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Leptin is essential for energy homeostasis and regulation of food intake. Patients with congenital generalized lipodystrophy (CGL) due to mutations in 1-acylglycerol-3-phosphate-O-acyltransferase 2 (AGPAT2) and the CGL murine model (Agpat2(-/-) mice) both have severe insulin resistance, diabetes mellitus, hepatic steatosis, and low plasma leptin levels. In this study, we show that continuous leptin treatment of Agpat2(-/-) mice for 28 days reduced plasma insulin and glucose levels and normalized hepatic steatosis and hypertriglyceridemia. Leptin also partially, but significantly, reversed the low plasma thyroxine and high corticosterone levels found in Agpat2(-/-) mice. Levels of carbohydrate response element binding protein (ChREBP) were reduced, whereas lipogenic gene expression were increased in the livers of Agpat2(-/-) mice, suggesting that deregulated ChREBP contributed to the development of fatty livers in these mice and that this transcription factor is a target of leptin's beneficial metabolic action. Leptin administration did not change hepatic fatty acid oxidation enzymes mRNA levels in Agpat2(-/-) mice. The selective deletion of leptin receptors only in hepatocytes did not prevent the positive metabolic actions of leptin in Agpat2(-/-) mice, supporting the notion that the majority of metabolic actions of leptin are dependent on its action in nonhepatocyte cells and/or the central nervous system.
Collapse
Affiliation(s)
- Víctor A Cortés
- Department of Molecular Genetics, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | | | | | | | | | | |
Collapse
|
48
|
Iizuka K, Wu W, Horikawa Y, Saito M, Takeda J. Feedback looping between ChREBP and PPARα in the regulation of lipid metabolism in brown adipose tissues. Endocr J 2013; 60:1145-53. [PMID: 23831548 DOI: 10.1507/endocrj.ej13-0079] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Carbohydrate response element binding protein (ChREBP) and peroxisome proliferator-activated receptor alpha (PPARα) play an important role in the regulation of lipid metabolism in the liver. Chrebp and Ppara mRNA levels are equally abundant in brown adipose tissue and liver. However, their functions in brown adipose tissues are unclear. In this study, we attempted to clarify the role of ChREBP and PPARα using brown adipose HB2 cell lines and tissues from wild type and Chrebp-/- C57BL/6J mice. In liver and brown adipose tissues, Chrebpb mRNA levels in the fasting state were much lower than those fed ad libitum, while Ppara mRNA levels in the fasting state were much higher than in the fed state. In differentiated brown adipose HB2 cell lines, glucose increased mRNA levels of ChREBP target genes such as Chrebpb, Fasn, and Glut4 in a dose dependent manner, while glucose decreased both Chrebpa and Ppara mRNA levels. Accordingly, adenoviral overexpression of ChREBP and a reporter assay demonstrated that ChREBP partially suppressed Ppara and Acox mRNA expression. Moreover, in brown adipose tissues from Chrebp-/- mice, Chrebpb and Fasn mRNA levels in the ad libitum fed state were much lower than those in the fasting state, while Ppara and Acox mRNA levels were not. Finally, using Wy14,643, a selective PPARα agonist, and overexpression of PPARα partially suppressed glucose induction of Chrebpb and Fasn mRNA in HB2 cells. In conclusion, the feedback loop between ChREBP and PPARα plays an important role in the regulation of lipogenesis in brown adipocytes.
Collapse
Affiliation(s)
- Katsumi Iizuka
- Department of Diabetes and Endocrinology, Graduate School of Medicine, Gifu University, Gifu, Japan
| | | | | | | | | |
Collapse
|