1
|
Fischer Z, Nauman C, Shayestehpour S, Pence L, Bouyain S, Yao X, Dobens LL. The Drosophila pseudokinase Tribbles translocates to the fat body membrane in response to fasting to modulate insulin sensitivity. Development 2025; 152:dev204493. [PMID: 40292740 PMCID: PMC12070071 DOI: 10.1242/dev.204493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 04/30/2025]
Abstract
The Drosophila pseudokinase Tribbles (Trbl) shares conserved functions with human TRIB3 to bind and inhibit Akt phosphorylation-activation by the Insulin Receptor (InR) to reduce insulin responses; consistent with this, increased levels of human TRIB3 are linked to type 2 diabetes. Here, we show that in fat body cells of well-fed Drosophila larvae, Trbl expression is low and predominantly in the nucleus while fasting or genetic reduction of insulin signaling resulted in increased Trbl expression and Trbl protein translocation to the plasma membrane. An E/G mutation in the Trbl pseudokinase kinase activation loop dominantly interfered with Trbl function leading to increased Akt activity, increased stability of Trbl substrates, including Trbl itself, and aberrant redistribution of Trbl multimers to the membrane. Several strategies designed to increase Akt activity were sufficient to translocate Trbl to the membrane, consistent with the notion that subcellular trafficking of Trbl to the fat body cell membrane acts a rheostat to reduce the strength of Akt-mediated insulin responses, counter to the InR, which has been shown to redistribute away from the membrane to modulate insulin signaling.
Collapse
Affiliation(s)
- Zachary Fischer
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Christopher Nauman
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Shima Shayestehpour
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Laramie Pence
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Samuel Bouyain
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Xiaolan Yao
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Leonard L. Dobens
- Division of Biology and Biomedical Engineering, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| |
Collapse
|
2
|
Chaudhary R, Weiskirchen R, Ehrlich M, Henis YI. Dual signaling pathways of TGF-β superfamily cytokines in hepatocytes: balancing liver homeostasis and disease progression. Front Pharmacol 2025; 16:1580500. [PMID: 40260391 PMCID: PMC12009898 DOI: 10.3389/fphar.2025.1580500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/23/2025] Open
Abstract
The transforming growth factor-β (TGF-β) superfamily (TGF-β-SF) comprises over 30 cytokines, including TGF-β, activins/inhibins, bone morphogenetic proteins (BMPs), and growth differentiation factors (GDFs). These cytokines play critical roles in liver function and disease progression. Here, we discuss Smad-dependent (canonical) and non-Smad pathways activated by these cytokines in a hepatocellular context. We highlight the connection between the deregulation of these pathways or the balance between them and key hepatocellular processes (e.g., proliferation, apoptosis, and epithelial-mesenchymal transition (EMT)). We further discuss their contribution to various chronic liver conditions, such as metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), and hepatocellular carcinoma (HCC). In MASLD and MASH, TGF-β signaling contributes to hepatocyte lipid accumulation, cell death and fibrosis progression through both Smad and non-Smad pathways. In HCC, TGF-β and other TGF-β-SF cytokines have a dual role, acting as tumor suppressors or promoters in early vs. advanced stages of tumor progression, respectively. Additionally, we review the involvement of non-Smad pathways in modulating hepatocyte responses to TGF-β-SF cytokines, particularly in the context of chronic liver diseases, as well as the interdependence with other key pathways (cholesterol metabolism, insulin resistance, oxidative stress and lipotoxicity) in MASLD/MASH pathogenesis. The perspectives and insights detailed in this review may assist in determining future research directions and therapeutic targets in liver conditions, including chronic liver diseases and cancer.
Collapse
Affiliation(s)
- Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Aachen, Germany
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yoav I. Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Meng TK, Han RL, Ma P, Chen SX, Qi BH, Wang ZX, Li XY, Deng HS. Microemulsion-based drug delivery system identifies pepper alkaloids as anti-obesity compounds. Acta Pharmacol Sin 2025:10.1038/s41401-025-01521-x. [PMID: 40113987 DOI: 10.1038/s41401-025-01521-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/23/2025] [Indexed: 03/22/2025]
Abstract
Obesity is a significant contributor to various metabolic diseases such as heart disease and diabetes. Due to the adverse effects of synthetic anti-obesity drugs, natural products from functional food plants, which mimic the effects of synthetic chemicals, present promising alternatives. However, many natural plant-derived compounds are poorly soluble in water, resulting in low bioavailability within the gastrointestinal tract, a key limitation for the effectiveness of many hydrophobic substances. In this study we developed a microemulsion-based drug delivery system in Drosophila, which effectively enhanced the solubility of hydrophobic compounds without noticeable effects on food intake or survival in fruit flies. This system consisted of cremophor EL, ethanol and ethyl oleate (7:6:1), which enabled the establishment of an emulsion-based liquid high-fat diet (LHFD) model, followed by a pilot screening of 161 standard substances from traditional Chinese medicine. We found that piperine (PIP), an alkaloid derived from black pepper, significantly decreased triacylglycerol (TAG) levels in both the intestine and in whole flies. We demonstrated that piperine (1 mg/ml) significantly elevated cytosolic Ca2+ levels in enterocytes by activating Transient receptor potential (TRP) channels. TRPV1 agonists such as capsaicin and evodiamine (another alkaloid identified during the screening) also exhibited anti-obesity effects. Increased Ca2+ levels resulted in the suppression of dietary lipase Magro expression through the activation of the transcription factor cAMP response element binding protein (CREB). Furthermore, hydrophobic compounds in the microemulsion were successfully delivered to distal tissues including liver and brain blood vessels in mice, and PIP in the microemulsion was sufficient to reduce body weight in mice. In conclusion, we have developed a microemulsion-based U-GLAD platform for drug delivery, and piperine is identified as a weight-controlling compound, providing a novel approach to the treatment of obesity and its associated symptoms.
Collapse
Affiliation(s)
- Tian-Kai Meng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Ruo-Lei Han
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Peng Ma
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shu-Xin Chen
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bo-Han Qi
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zi-Xuan Wang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiao-Yu Li
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Han-Song Deng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
4
|
Singh MK, Ryu TH, Nguyen MN, Yu K. Inhibition of high-fat diet-induced miRNA ameliorates tau toxicity in Drosophila. Biochem Biophys Res Commun 2024; 733:150446. [PMID: 39067249 DOI: 10.1016/j.bbrc.2024.150446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Alzheimer's disease (AD), caused by amyloid beta (Aβ) plaques and Tau tangles, is a neurodegenerative disease characterized by progressive memory impairment and cognitive dysfunction. High-fat diet (HFD), which induces type 2 diabetes, exacerbates Aβ plaque deposition in the brain. To investigate the function of HFD in Tau-mediated AD, we fed an HFD to the Drosophila Tau model and found that HFD aggravates Tau-induced neurological phenotypes. Since microRNAs (miRNAs) are biomarkers for diabetes and AD, we evaluated the expression levels of common miRNAs of HFD and AD in HFD-fed Tau model fly brains. Among the common miRNAs, the expression levels of Let-7 and miR-34 were increased. We found that the inhibition of these miRNAs alleviates Tau-mediated AD phenotypes. Our research provides valuable insights into how HFD accelerates tau toxicity. Additionally, our work highlights the therapeutic potential of targeting Let-7 and miR-34 to develop innovative treatment approaches for AD.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Tae Hoon Ryu
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Minh Nguyet Nguyen
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Bioscience, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Kweon Yu
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Department of Bioscience, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
5
|
Rundell TB, Baranski TJ. Insect Models to Study Human Lipid Metabolism Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39405006 DOI: 10.1007/5584_2024_827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Disorders of lipid metabolism such as obesity have become some of the most significant diseases of the twenty-first century. Despite these metabolic diseases affecting more than a third of the population in highly industrialized nations, the mechanisms underlying disease development remain poorly understood. Insect models, such as Drosophila melanogaster, offer a means of systematically examining conserved lipid metabolism and its pathology. Over the past several decades, Drosophila melanogaster has been used to greatly expand on our knowledge of metabolic disease, often taking advantage of the extensive genetic tools available to researchers. Additionally, Drosophila melanogaster has served and will continue to serve as a powerful tool for validating the results of genome-wide approaches to the study of diseases. This chapter explores the advancements of insect models in the study of lipid metabolism disorders as well as highlight opportunities for future areas of research.
Collapse
Affiliation(s)
- Thomas B Rundell
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Thomas J Baranski
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
6
|
Akiyama T, Raftery LA, Wharton KA. Bone morphogenetic protein signaling: the pathway and its regulation. Genetics 2024; 226:iyad200. [PMID: 38124338 PMCID: PMC10847725 DOI: 10.1093/genetics/iyad200] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/27/2023] [Indexed: 12/23/2023] Open
Abstract
In the mid-1960s, bone morphogenetic proteins (BMPs) were first identified in the extracts of bone to have the remarkable ability to induce heterotopic bone. When the Drosophila gene decapentaplegic (dpp) was first identified to share sequence similarity with mammalian BMP2/BMP4 in the late-1980s, it became clear that secreted BMP ligands can mediate processes other than bone formation. Following this discovery, collaborative efforts between Drosophila geneticists and mammalian biochemists made use of the strengths of their respective model systems to identify BMP signaling components and delineate the pathway. The ability to conduct genetic modifier screens in Drosophila with relative ease was critical in identifying the intracellular signal transducers for BMP signaling and the related transforming growth factor-beta/activin signaling pathway. Such screens also revealed a host of genes that encode other core signaling components and regulators of the pathway. In this review, we provide a historical account of this exciting time of gene discovery and discuss how the field has advanced over the past 30 years. We have learned that while the core BMP pathway is quite simple, composed of 3 components (ligand, receptor, and signal transducer), behind the versatility of this pathway lies multiple layers of regulation that ensures precise tissue-specific signaling output. We provide a sampling of these discoveries and highlight many questions that remain to be answered to fully understand the complexity of BMP signaling.
Collapse
Affiliation(s)
- Takuya Akiyama
- Department of Biology, Rich and Robin Porter Cancer Research Center, The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Laurel A Raftery
- School of Life Sciences, University of Nevada, 4505 S. Maryland Parkway, Las Vegas, NV 89154, USA
| | - Kristi A Wharton
- Department of Molecular Biology, Cell Biology, and Biochemistry, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| |
Collapse
|
7
|
Liu X, Cai YD, Chiu JC. Regulation of protein O-GlcNAcylation by circadian, metabolic, and cellular signals. J Biol Chem 2024; 300:105616. [PMID: 38159854 PMCID: PMC10810748 DOI: 10.1016/j.jbc.2023.105616] [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: 07/24/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAcylation) is a dynamic post-translational modification that regulates thousands of proteins and almost all cellular processes. Aberrant O-GlcNAcylation has been associated with numerous diseases, including cancer, neurodegenerative diseases, cardiovascular diseases, and type 2 diabetes. O-GlcNAcylation is highly nutrient-sensitive since it is dependent on UDP-GlcNAc, the end product of the hexosamine biosynthetic pathway (HBP). We previously observed daily rhythmicity of protein O-GlcNAcylation in a Drosophila model that is sensitive to the timing of food consumption. We showed that the circadian clock is pivotal in regulating daily O-GlcNAcylation rhythms given its control of the feeding-fasting cycle and hence nutrient availability. Interestingly, we reported that the circadian clock also modulates daily O-GlcNAcylation rhythm by regulating molecular mechanisms beyond the regulation of food consumption time. A large body of work now indicates that O-GlcNAcylation is likely a generalized cellular status effector as it responds to various cellular signals and conditions, such as ER stress, apoptosis, and infection. In this review, we summarize the metabolic regulation of protein O-GlcNAcylation through nutrient availability, HBP enzymes, and O-GlcNAc processing enzymes. We discuss the emerging roles of circadian clocks in regulating daily O-GlcNAcylation rhythm. Finally, we provide an overview of other cellular signals or conditions that impact O-GlcNAcylation. Many of these cellular pathways are themselves regulated by the clock and/or metabolism. Our review highlights the importance of maintaining optimal O-GlcNAc rhythm by restricting eating activity to the active period under physiological conditions and provides insights into potential therapeutic targets of O-GlcNAc homeostasis under pathological conditions.
Collapse
Affiliation(s)
- Xianhui Liu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Yao D Cai
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Joanna C Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA.
| |
Collapse
|
8
|
Škrgat S, Harlander M, Janić M. Obesity and Insulin Resistance in Asthma Pathogenesis and Clinical Outcomes. Biomedicines 2024; 12:173. [PMID: 38255279 PMCID: PMC10813771 DOI: 10.3390/biomedicines12010173] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Common inflammatory ground links obesity, insulin resistance, and asthma. As recognition of their interplay, one worsening the natural course of the other, is recognised, questions remain about how to adequately address them altogether to improve clinical outcomes. The present manuscript sheds light on the problem, describing possible pathophysiological links, clinical views, and therapeutic challenges, raising questions about what remains to be done, and calling for multidisciplinary treatment of these patients to detect diseases early and adequately address them before they become full-blown and deteriorate their health and quality of life.
Collapse
Affiliation(s)
- Sabina Škrgat
- Department of Pulmonary Diseases and Allergy, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
- Medical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Matevž Harlander
- Department of Pulmonary Diseases and Allergy, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia;
- Medical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | - Miodrag Janić
- Medical Faculty, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
- Clinical Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
9
|
Jia X, Chen Q, Wu H, Liu H, Jing C, Gong A, Zhang Y. Exploring a novel therapeutic strategy: the interplay between gut microbiota and high-fat diet in the pathogenesis of metabolic disorders. Front Nutr 2023; 10:1291853. [PMID: 38192650 PMCID: PMC10773723 DOI: 10.3389/fnut.2023.1291853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
In the past two decades, the rapid increase in the incidence of metabolic diseases, including obesity, diabetes, dyslipidemia, non-alcoholic fatty liver disease, hypertension, and hyperuricemia, has been attributed to high-fat diets (HFD) and decreased physical activity levels. Although the phenotypes and pathologies of these metabolic diseases vary, patients with these diseases exhibit disease-specific alterations in the composition and function of their gut microbiota. Studies in germ-free mice have shown that both HFD and gut microbiota can promote the development of metabolic diseases, and HFD can disrupt the balance of gut microbiota. Therefore, investigating the interaction between gut microbiota and HFD in the pathogenesis of metabolic diseases is crucial for identifying novel therapeutic strategies for these diseases. This review takes HFD as the starting point, providing a detailed analysis of the pivotal role of HFD in the development of metabolic disorders. It comprehensively elucidates the impact of HFD on the balance of intestinal microbiota, analyzes the mechanisms underlying gut microbiota dysbiosis leading to metabolic disruptions, and explores the associated genetic factors. Finally, the potential of targeting the gut microbiota as a means to address metabolic disturbances induced by HFD is discussed. In summary, this review offers theoretical support and proposes new research avenues for investigating the role of nutrition-related factors in the pathogenesis of metabolic disorders in the organism.
Collapse
Affiliation(s)
- Xiaokang Jia
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huiwen Wu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Hongbo Liu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Chunying Jing
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Aimin Gong
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Yuanyuan Zhang
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Bakopoulos D, Golenkina S, Dark C, Christie EL, Sánchez-Sánchez BJ, Stramer BM, Cheng LY. Convergent insulin and TGF-β signalling drives cancer cachexia by promoting aberrant fat body ECM accumulation in a Drosophila tumour model. EMBO Rep 2023; 24:e57695. [PMID: 38014610 DOI: 10.15252/embr.202357695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Abstract
In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-β signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-β signalling to regulate ECM accumulation in the fat body. TGF-β signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-β signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.
Collapse
Affiliation(s)
- Daniel Bakopoulos
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Brian M Stramer
- Kings College London, Randall Centre for Cell & Molecular Biophysics, London, UK
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
11
|
Kuroiwa T, Lui H, Nakagawa K, Iida N, Desrochers C, Wan R, Adam E, Larson D, Amadio P, Gingery A. Impact of High Fat Diet and Sex in a Rabbit Model of Carpal Tunnel Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549152. [PMID: 37546859 PMCID: PMC10402177 DOI: 10.1101/2023.07.15.549152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Carpal tunnel syndrome (CTS) is a common musculoskeletal disorder, characterized by fibrosis of the subsynovial connective tissue (SSCT) mediated by transforming growth factor beta (TGF-β). Risk factors for CTS include metabolic dysfunction and age. Additionally, the incidence of CTS is higher in women. In this study we hypothesized that a high-fat diet (HFD), a common driver of metabolic dysfunction, would promote SSCT fibrosis found in CTS and that this response would be sex dependent. To test this, we examined the effects of HFD and sex on SSCT fibrosis using our established rabbit model of CTS. Forty-eight (24 male, 24 female) adult rabbits were divided into four groups including HFD or standard diet with and without CTS induction. SSCT was collected for histological and gene expression analysis. HFD promoted SSCT thickening and upregulated profibrotic genes, including TGF-β. Fibrotic genes were differentially expressed in males and females. Interestingly while the prevalence of CTS is greater in women than in men, the converse is observed in the presence of metabolic dysfunction. This work recapitulates this clinical observation and begins to elucidate the sex-based differences found in SSCT fibrosis. This knowledge should drive further research and may lead to metabolic and sex specific therapeutic strategies for the treatment of patients with CTS.
Collapse
|
12
|
Qian W, Guo M, Peng J, Zhao T, Li Z, Yang Y, Li H, Zhang X, King-Jones K, Cheng D. Decapentaplegic retards lipolysis during metamorphosis in Bombyx mori and Drosophila melanogaster. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 155:103928. [PMID: 36870515 DOI: 10.1016/j.ibmb.2023.103928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/29/2023] [Accepted: 03/01/2023] [Indexed: 05/10/2023]
Abstract
Insect morphogen decapentaplegic (Dpp) functions as one of the key extracellular ligands of the Bone Morphogenetic Protein (BMP) signaling pathway. Previous studies in insects mainly focused on the roles of Dpp during embryonic development and the formation of adult wings. In this study, we demonstrate a new role for Dpp in retarding lipolysis during metamorphosis in both Bombyx mori and Drosophila melanogaster. CRISPR/Cas9-mediated mutation of Bombyx dpp causes pupal lethality, induces an excessive and premature breakdown of lipids in the fat body, and upregulates the expressions of several lipolytic enzyme genes, including brummer (bmm), lipase 3 (lip3), and hormone-sensitive lipase (hsl), and lipid storage droplet 1 (lsd1), a lipid droplets (LD)-associated protein gene. Further investigation in Drosophila reveals that salivary gland-specific knockdown of the dpp gene and fat body-specific knockdown of Mad involved in Dpp signaling phenocopy the effects of Bombyx dpp mutation on pupal development and lipolysis. Taken together, our data indicate that the Dpp-mediated BMP signaling in the fat body maintains lipid homeostasis by retarding lipolysis, which is necessary for pupa-adult transition during insect metamorphosis.
Collapse
Affiliation(s)
- Wenliang Qian
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Mengge Guo
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Jian Peng
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Tujing Zhao
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Zheng Li
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Yan Yang
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Hao Li
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Xing Zhang
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Kirst King-Jones
- Department of Biological Sciences, University of Alberta, G-504 Biological Sciences Bldg., Edmonton, Alberta, T6G 2E9, Canada.
| | - Daojun Cheng
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
13
|
Popovic R, Yu Y, Leal NS, Fedele G, Loh SHY, Martins LM. Upregulation of Tribbles decreases body weight and increases sleep duration. Dis Model Mech 2023; 16:dmm049942. [PMID: 37083954 PMCID: PMC10151826 DOI: 10.1242/dmm.049942] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 03/16/2023] [Indexed: 04/22/2023] Open
Abstract
Eukaryotic Tribbles proteins are pseudoenzymes that regulate multiple aspects of intracellular signalling. Both Drosophila melanogaster and mammalian members of this family of pseudokinases act as negative regulators of insulin signalling. Mammalian tribbles pseudokinase (TRIB) genes have also been linked to insulin resistance and type 2 diabetes mellitus. Type 2 diabetes mellitus is associated with increased body weight, sleep problems and increased long-term mortality. Here, we investigated how manipulating the expression of Tribbles impacts body weight, sleep and mortality. We showed that the overexpression of Drosophila tribbles (trbl) in the fly fat body reduces both body weight and lifespan in adult flies without affecting food intake. Furthermore, it decreases the levels of Drosophila insulin-like peptide 2 (DILP2; ILP2) and increases night-time sleep. The three genes encoding TRIBs of mammals, TRIB1, TRIB2 and TRIB3, show both common and unique features. As the three human TRIB genes share features with Drosophila trbl, we further explored the links between TRIB genetic variants and both body weight and sleep in the human population. We identified associations between the polymorphisms and expression levels of the pseudokinases and markers of body weight and sleep duration. We conclude that Tribbles pseudokinases are involved in the control of body weight, lifespan and sleep.
Collapse
Affiliation(s)
- Rebeka Popovic
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Nuno Santos Leal
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Giorgio Fedele
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Samantha H. Y. Loh
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - L. Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
14
|
The Role of Gut Microbiota in High-Fat-Diet-Induced Diabetes: Lessons from Animal Models and Humans. Nutrients 2023; 15:nu15040922. [PMID: 36839280 PMCID: PMC9963658 DOI: 10.3390/nu15040922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
The number of diabetes mellitus patients is increasing rapidly worldwide. Diet and nutrition are strongly believed to play a significant role in the development of diabetes mellitus. However, the specific dietary factors and detailed mechanisms of its development have not been clearly elucidated. Increasing evidence indicates the intestinal microbiota is becoming abundantly apparent in the progression and prevention of insulin resistance in diabetes. Differences in gut microbiota composition, particularly butyrate-producing bacteria, have been observed in preclinical animal models as well as human patients compared to healthy controls. Gut microbiota dysbiosis may disrupt intestinal barrier functions and alter host metabolic pathways, directly or indirectly relating to insulin resistance. In this article, we focus on dietary fat, diabetes, and gut microbiome characterization. The promising probiotic and prebiotic approaches to diabetes, by favorably modifying the composition of the gut microbial community, warrant further investigation through well-designed human clinical studies.
Collapse
|
15
|
Tsao DD, Chang KR, Kockel L, Park S, Kim SK. A genetic strategy to measure insulin signaling regulation and physiology in Drosophila. PLoS Genet 2023; 19:e1010619. [PMID: 36730473 PMCID: PMC9928101 DOI: 10.1371/journal.pgen.1010619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 02/14/2023] [Accepted: 01/15/2023] [Indexed: 02/04/2023] Open
Abstract
Insulin regulation is a hallmark of health, and impaired insulin signaling promotes metabolic diseases like diabetes mellitus. However, current assays for measuring insulin signaling in all animals remain semi-quantitative and lack the sensitivity, tissue-specificity or temporal resolution needed to quantify in vivo physiological signaling dynamics. Insulin signal transduction is remarkably conserved across metazoans, including insulin-dependent phosphorylation and regulation of Akt/Protein kinase B. Here, we generated transgenic fruit flies permitting tissue-specific expression of an immunoepitope-labelled Akt (AktHF). We developed enzyme-linked immunosorption assays (ELISA) to quantify picomolar levels of phosphorylated (pAktHF) and total AktHF in single flies, revealing dynamic tissue-specific physiological regulation of pAktHF in response to fasting and re-feeding, exogenous insulin, or targeted genetic suppression of established insulin signaling regulators. Genetic screening revealed Pp1-87B as an unrecognized regulator of Akt and insulin signaling. Tools and concepts here provide opportunities to discover tissue-specific regulators of in vivo insulin signaling responses.
Collapse
Affiliation(s)
- Deborah D. Tsao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Kathleen R. Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lutz Kockel
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Sangbin Park
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine (Division of Endocrinology, Metabolism, Gerontology), Stanford University School of Medicine, Stanford, California, United States of America
- Department of Pediatrics (Division of Endocrinology), Stanford University School of Medicine, Stanford, California, United States of America
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
16
|
Shang S, Liu J, Hua F. Protein acylation: mechanisms, biological functions and therapeutic targets. Signal Transduct Target Ther 2022; 7:396. [PMID: 36577755 PMCID: PMC9797573 DOI: 10.1038/s41392-022-01245-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/27/2022] [Accepted: 11/06/2022] [Indexed: 12/30/2022] Open
Abstract
Metabolic reprogramming is involved in the pathogenesis of not only cancers but also neurodegenerative diseases, cardiovascular diseases, and infectious diseases. With the progress of metabonomics and proteomics, metabolites have been found to affect protein acylations through providing acyl groups or changing the activities of acyltransferases or deacylases. Reciprocally, protein acylation is involved in key cellular processes relevant to physiology and diseases, such as protein stability, protein subcellular localization, enzyme activity, transcriptional activity, protein-protein interactions and protein-DNA interactions. Herein, we summarize the functional diversity and mechanisms of eight kinds of nonhistone protein acylations in the physiological processes and progression of several diseases. We also highlight the recent progress in the development of inhibitors for acyltransferase, deacylase, and acylation reader proteins for their potential applications in drug discovery.
Collapse
Affiliation(s)
- Shuang Shang
- grid.506261.60000 0001 0706 7839CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050 Beijing, P.R. China
| | - Jing Liu
- grid.506261.60000 0001 0706 7839CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050 Beijing, P.R. China
| | - Fang Hua
- grid.506261.60000 0001 0706 7839CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050 Beijing, P.R. China
| |
Collapse
|
17
|
Utility of Hypoglycemic Agents to Treat Asthma with Comorbid Obesity. Pulm Ther 2022; 9:71-89. [PMID: 36575356 PMCID: PMC9931991 DOI: 10.1007/s41030-022-00211-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/08/2022] [Indexed: 12/29/2022] Open
Abstract
Adults with obesity may develop asthma that is ineffectively controlled by inhaled corticosteroids and long-acting beta-adrenoceptor agonists. Mechanistic and translational studies suggest that metabolic dysregulation that occurs with obesity, particularly hyperglycemia and insulin resistance, contributes to altered immune cell function and low-grade systemic inflammation. Importantly, in these cases, the same proinflammatory cytokines believed to contribute to insulin resistance may also be responsible for airway remodeling and hyperresponsiveness. In the past decade, new research has emerged assessing whether hypoglycemic therapies impact comorbid asthma as reflected by the incidence of asthma, asthma-related emergency department visits, asthma-related hospitalizations, and asthma-related exacerbations. The purpose of this review article is to discuss the mechanism of action, preclinical data, and existing clinical studies regarding the efficacy and safety of hypoglycemic therapies for adults with obesity and comorbid asthma.
Collapse
|
18
|
Circulating microRNAs from early childhood and adolescence are associated with pre-diabetes at 18 years of age in women from the PMNS cohort. J Dev Orig Health Dis 2022; 13:806-811. [PMID: 35450554 DOI: 10.1017/s2040174422000137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
With type 2 diabetes presenting at younger ages, there is a growing need to identify biomarkers of future glucose intolerance. A high (20%) prevalence of glucose intolerance at 18 years was seen in women from the Pune Maternal Nutrition Study (PMNS) birth cohort. We investigated the potential of circulating microRNAs in risk stratification for future pre-diabetes in these women. Here, we provide preliminary longitudinal analyses of circulating microRNAs in normal glucose tolerant (NGT@18y, N = 10) and glucose intolerant (N = 8) women (ADA criteria) at 6, 12 and 17 years of their age using discovery analysis (OpenArray™ platform). Machine-learning workflows involving Lasso with bootstrapping/leave-one-out cross-validation identified microRNAs associated with glucose intolerance at 18 years of age. Several microRNAs, including miR-212-3p, miR-30e-3p and miR-638, stratified glucose-intolerant women from NGT at childhood. Our results suggest that circulating microRNAs, longitudinally assessed over 17 years of life, are dynamic biomarkers associated with and predictive of pre-diabetes at 18 years of age. Validation of these findings in males and remaining participants from the PMNS birth cohort will provide a unique opportunity to study novel epigenetic mechanisms in the life-course progression of glucose intolerance and enhance current clinical risk prediction of pre-diabetes and progression to type 2 diabetes.
Collapse
|
19
|
Elmas O, Cenik P, Sirinyildiz F, Elmas S, Sirin F, Cesur G. Relationship between cognitive functions, levels of NR2A
and NR2B subunits of hippocampal NMDA receptors, serum
TGF-β1 level, and oxidative stress in rats fed a high-fat diet. JOURNAL OF ANIMAL AND FEED SCIENCES 2022. [DOI: 10.22358/jafs/152027/2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
20
|
Metabolic Syndrome: Lessons from Rodent and Drosophila Models. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5850507. [PMID: 35782067 PMCID: PMC9242782 DOI: 10.1155/2022/5850507] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/20/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022]
Abstract
Overweight and obesity are health conditions tightly related to a number of metabolic complications collectively called “metabolic syndrome” (MetS). Clinical diagnosis of MetS includes the presence of the increased waist circumference or so-called abdominal obesity, reduced high density lipoprotein level, elevated blood pressure, and increased blood glucose and triacylglyceride levels. Different approaches, including diet-induced and genetically induced animal models, have been developed to study MetS pathogenesis and underlying mechanisms. Studies of metabolic disturbances in the fruit fly Drosophila and mammalian models along with humans have demonstrated that fruit flies and small mammalian models like rats and mice have many similarities with humans in basic metabolic functions and share many molecular mechanisms which regulate these metabolic processes. In this paper, we describe diet-induced, chemically and genetically induced animal models of the MetS. The advantages and limitations of rodent and Drosophila models of MetS and obesity are also analyzed.
Collapse
|
21
|
Salas-Venegas V, Flores-Torres RP, Rodríguez-Cortés YM, Rodríguez-Retana D, Ramírez-Carreto RJ, Concepción-Carrillo LE, Pérez-Flores LJ, Alarcón-Aguilar A, López-Díazguerrero NE, Gómez-González B, Chavarría A, Konigsberg M. The Obese Brain: Mechanisms of Systemic and Local Inflammation, and Interventions to Reverse the Cognitive Deficit. Front Integr Neurosci 2022; 16:798995. [PMID: 35422689 PMCID: PMC9002268 DOI: 10.3389/fnint.2022.798995] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Overweight and obesity are now considered a worldwide pandemic and a growing public health problem with severe economic and social consequences. Adipose tissue is an organ with neuroimmune-endocrine functions, which participates in homeostasis. So, adipocyte hypertrophy and hyperplasia induce a state of chronic inflammation that causes changes in the brain and induce neuroinflammation. Studies with obese animal models and obese patients have shown a relationship between diet and cognitive decline, especially working memory and learning deficiencies. Here we analyze how obesity-related peripheral inflammation can affect central nervous system physiology, generating neuroinflammation. Given that the blood-brain barrier is an interface between the periphery and the central nervous system, its altered physiology in obesity may mediate the consequences on various cognitive processes. Finally, several interventions, and the use of natural compounds and exercise to prevent the adverse effects of obesity in the brain are also discussed.
Collapse
Affiliation(s)
- Verónica Salas-Venegas
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Rosa Pamela Flores-Torres
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Yesica María Rodríguez-Cortés
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Diego Rodríguez-Retana
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Ricardo Jair Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Luis Edgar Concepción-Carrillo
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Laura Josefina Pérez-Flores
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Norma Edith López-Díazguerrero
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Beatriz Gómez-González
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
- *Correspondence: Mina Konigsberg,
| |
Collapse
|
22
|
Havula E, Ghazanfar S, Lamichane N, Francis D, Hasygar K, Liu Y, Alton LA, Johnstone J, Needham EJ, Pulpitel T, Clark T, Niranjan HN, Shang V, Tong V, Jiwnani N, Audia G, Alves AN, Sylow L, Mirth C, Neely GG, Yang J, Hietakangas V, Simpson SJ, Senior AM. Genetic variation of macronutrient tolerance in Drosophila melanogaster. Nat Commun 2022; 13:1637. [PMID: 35347148 PMCID: PMC8960806 DOI: 10.1038/s41467-022-29183-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 02/28/2022] [Indexed: 11/08/2022] Open
Abstract
Carbohydrates, proteins and lipids are essential nutrients to all animals; however, closely related species, populations, and individuals can display dramatic variation in diet. Here we explore the variation in macronutrient tolerance in Drosophila melanogaster using the Drosophila genetic reference panel, a collection of ~200 strains derived from a single natural population. Our study demonstrates that D. melanogaster, often considered a "dietary generalist", displays marked genetic variation in survival on different diets, notably on high-sugar diet. Our genetic analysis and functional validation identify several regulators of macronutrient tolerance, including CG10960/GLUT8, Pkn and Eip75B. We also demonstrate a role for the JNK pathway in sugar tolerance and de novo lipogenesis. Finally, we report a role for tailless, a conserved orphan nuclear hormone receptor, in regulating sugar metabolism via insulin-like peptide secretion and sugar-responsive CCHamide-2 expression. Our study provides support for the use of nutrigenomics in the development of personalized nutrition.
Collapse
Affiliation(s)
- E Havula
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - S Ghazanfar
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - N Lamichane
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - D Francis
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - K Hasygar
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Y Liu
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - L A Alton
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - J Johnstone
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - E J Needham
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - T Pulpitel
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - T Clark
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - H N Niranjan
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Shang
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Tong
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - N Jiwnani
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - G Audia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - A N Alves
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - L Sylow
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - C Mirth
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - G G Neely
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - J Yang
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - V Hietakangas
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - S J Simpson
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - A M Senior
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
23
|
Guo X, Sunil C, Qian G. Obesity and the Development of Lung Fibrosis. Front Pharmacol 2022; 12:812166. [PMID: 35082682 PMCID: PMC8784552 DOI: 10.3389/fphar.2021.812166] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Obesity is an epidemic worldwide and the obese people suffer from a range of respiratory complications including fibrotic changes in the lung. The influence of obesity on the lung is multi-factorial, which is related to both mechanical injury and various inflammatory mediators produced by excessive adipose tissues, and infiltrated immune cells. Adiposity causes increased production of inflammatory mediators, for example, cytokines, chemokines, and adipokines, both locally and in the systemic circulation, thereby rendering susceptibility to respiratory diseases, and altered responses. Lung fibrosis is closely related to chronic inflammation in the lung. Current data suggest a link between lung fibrosis and diet-induced obesity, although the mechanism remains incomplete understood. This review summarizes findings on the association of lung fibrosis with obesity, highlights the role of several critical inflammatory mediators (e.g., TNF-α, TGF-β, and MCP-1) in obesity related lung fibrosis and the implication of obesity in the outcomes of idiopathic pulmonary fibrosis patients.
Collapse
Affiliation(s)
- Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Christudas Sunil
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
24
|
He S, Ryu J, Liu J, Luo H, Lv Y, Langlais PR, Wen J, Dong F, Sun Z, Xia W, Lynch JL, Duggirala R, Nicholson BJ, Zang M, Shi Y, Zhang F, Liu F, Bai J, Dong LQ. LRG1 is an adipokine that mediates obesity-induced hepatosteatosis and insulin resistance. J Clin Invest 2021; 131:148545. [PMID: 34730111 DOI: 10.1172/jci148545] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Dysregulation in adipokine biosynthesis and function contributes to obesity-induced metabolic diseases. However, the identities and functions of many of the obesity-induced secretory molecules remain unknown. Here, we report the identification of leucine-rich alpha-2-glycoprotein 1 (LRG1) as an obesity-associated adipokine that exacerbates high fat diet-induced hepatosteatosis and insulin resistance. Serum levels of LRG1 were markedly elevated in obese humans and mice compared to their respective controls. LRG1 deficiency in mice greatly alleviated diet-induced hepatosteatosis, obesity, and insulin resistance. Mechanistically, LRG1 bound with high selectivity to the liver and promoted hepatosteatosis by increasing de novo lipogenesis and suppressing fatty acid β-oxidation. LRG1 also inhibited hepatic insulin signaling by down-regulating insulin receptor substrates 1 and 2. Our study identified LRG1 as a key molecule that mediates the crosstalk between adipocytes and hepatocytes in diet-induced hepatosteatosis and insulin resistance. Suppressing LRG1 expression and function may be a promising strategy for the treatment of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Sijia He
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Jiyoon Ryu
- Department of Cell Systems & Anatomy, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Juanhong Liu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hairong Luo
- Department of Metabolism and Endocrinology Clinical Research Center for Met, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Lv
- Novo Nordisk Research Centre China, Novo Nordisk Research Centre China, Beijing, China
| | - Paul R Langlais
- Department of Medicine, University of Arizona, Tucson, United States of America
| | - Jie Wen
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Dong
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Zhe Sun
- Novo Nordisk Research Centre China, Novo Nordisk Research Centre China, Beijing, China
| | - Wenjuan Xia
- Novo Nordisk Research Centre China, Novo Nordisk Research Centre China, Beijing, China
| | - Jane L Lynch
- Department of Pediatrics, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Ravindranath Duggirala
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, McAllen, United States of America
| | - Bruce J Nicholson
- Department of Biochemistry and Structural Biology, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Mengwei Zang
- Department of Molecular Medicine, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Yuguang Shi
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Fang Zhang
- Novo Nordisk Research Centre China, Novo Nordisk Research Centre China, Beijing, China
| | - Feng Liu
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Juli Bai
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, United States of America
| | - Lily Q Dong
- Department of Cellular and Structural Biology, University of Texas Health at San Antonio, San Antonio, United States of America
| |
Collapse
|
25
|
Sarangi M, Dus M. Crème de la Créature: Dietary Influences on Behavior in Animal Models. Front Behav Neurosci 2021; 15:746299. [PMID: 34658807 PMCID: PMC8511460 DOI: 10.3389/fnbeh.2021.746299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
In humans, alterations in cognitive, motivated, and affective behaviors have been described with consumption of processed diets high in refined sugars and saturated fats and with high body mass index, but the causes, mechanisms, and consequences of these changes remain poorly understood. Animal models have provided an opportunity to answer these questions and illuminate the ways in which diet composition, especially high-levels of added sugar and saturated fats, contribute to brain physiology, plasticity, and behavior. Here we review findings from invertebrate (flies) and vertebrate models (rodents, zebrafish) that implicate these diets with changes in multiple behaviors, including eating, learning and memory, and motivation, and discuss limitations, open questions, and future opportunities.
Collapse
Affiliation(s)
| | - Monica Dus
- Department of Molecular, Cellular, and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
26
|
Manna S, Karmakar P, Kisan B, Mishra M, Barooah N, Bhasikuttan AC, Mohanty J. Fibril-induced neurodegenerative disorders in an Aβ-mutant Drosophila model: therapeutic targeting using ammonium molybdate. Chem Commun (Camb) 2021; 57:8488-8491. [PMID: 34350921 DOI: 10.1039/d1cc03752h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ability of polyanionic molybdate to inhibit and degrade protein fibrils both in vitro (insulin protein) and in vivo (Drosophila fly model) has been demonstrated. We establish the disappearance of fibrillar structures and recovery from neurodegenerative disorders in molybdate-treated Aβ42-mutant Drosophila flies as compared to the untreated ones, corroborating the therapeutic ability of ammonium molybdate towards the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Sudipa Manna
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| | | | | | | | | | | | | |
Collapse
|
27
|
Tello-Flores VA, Beltrán-Anaya FO, Ramírez-Vargas MA, Esteban-Casales BE, Navarro-Tito N, Alarcón-Romero LDC, Luciano-Villa CA, Ramírez M, del Moral-Hernández Ó, Flores-Alfaro E. Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance. Int J Mol Sci 2021; 22:7256. [PMID: 34298896 PMCID: PMC8306787 DOI: 10.3390/ijms22147256] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are single-stranded RNA biomolecules with a length of >200 nt, and they are currently considered to be master regulators of many pathological processes. Recent publications have shown that lncRNAs play important roles in the pathogenesis and progression of insulin resistance (IR) and glucose homeostasis by regulating inflammatory and lipogenic processes. lncRNAs regulate gene expression by binding to other non-coding RNAs, mRNAs, proteins, and DNA. In recent years, several mechanisms have been reported to explain the key roles of lncRNAs in the development of IR, including metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), imprinted maternal-ly expressed transcript (H19), maternally expressed gene 3 (MEG3), myocardial infarction-associated transcript (MIAT), and steroid receptor RNA activator (SRA), HOX transcript antisense RNA (HOTAIR), and downregulated Expression-Related Hexose/Glucose Transport Enhancer (DREH). LncRNAs participate in the regulation of lipid and carbohydrate metabolism, the inflammatory process, and oxidative stress through different pathways, such as cyclic adenosine monophosphate/protein kinase A (cAMP/PKA), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), polypyrimidine tract-binding protein 1/element-binding transcription factor 1c (PTBP1/SREBP-1c), AKT/nitric oxide synthase (eNOS), AKT/forkhead box O1 (FoxO1), and tumor necrosis factor-alpha (TNF-α)/c-Jun-N-terminal kinases (JNK). On the other hand, the mechanisms linked to the molecular, cellular, and biochemical actions of lncRNAs vary according to the tissue, biological species, and the severity of IR. Therefore, it is essential to elucidate the role of lncRNAs in the insulin signaling pathway and glucose and lipid metabolism. This review analyzes the function and molecular mechanisms of lncRNAs involved in the development of IR.
Collapse
Affiliation(s)
- Vianet Argelia Tello-Flores
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Fredy Omar Beltrán-Anaya
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Marco Antonio Ramírez-Vargas
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Brenda Ely Esteban-Casales
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Luz del Carmen Alarcón-Romero
- Laboratorio de Citopatología e Histoquímica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Carlos Aldair Luciano-Villa
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Mónica Ramírez
- CONACyT, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Óscar del Moral-Hernández
- Laboratorio de Virología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
| | - Eugenia Flores-Alfaro
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| |
Collapse
|
28
|
Heier C, Klishch S, Stilbytska O, Semaniuk U, Lushchak O. The Drosophila model to interrogate triacylglycerol biology. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158924. [PMID: 33716135 DOI: 10.1016/j.bbalip.2021.158924] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022]
Abstract
The deposition of storage fat in the form of triacylglycerol (TAG) is an evolutionarily conserved strategy to cope with fluctuations in energy availability and metabolic stress. Organismal TAG storage in specialized adipose tissues provides animals a metabolic reserve that sustains survival during development and starvation. On the other hand, excessive accumulation of adipose TAG, defined as obesity, is associated with an increasing prevalence of human metabolic diseases. During the past decade, the fruit fly Drosophila melanogaster, traditionally used in genetics and developmental biology, has been established as a versatile model system to study TAG metabolism and the etiology of lipid-associated metabolic diseases. Similar to humans, Drosophila TAG homeostasis relies on the interplay of organ systems specialized in lipid uptake, synthesis, and processing, which are integrated by an endocrine network of hormones and messenger molecules. Enzymatic formation of TAG from sugar or dietary lipid, its storage in lipid droplets, and its mobilization by lipolysis occur via mechanisms largely conserved between Drosophila and humans. Notably, dysfunctional Drosophila TAG homeostasis occurs in the context of aging, overnutrition, or defective gene function, and entails tissue-specific and organismal pathologies that resemble human disease. In this review, we summarize the physiology and biochemistry of TAG in Drosophila and outline the potential of this organism as a model system to understand the genetic and dietary basis of TAG storage and TAG-related metabolic disorders.
Collapse
Affiliation(s)
- Christoph Heier
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Humboldtstrasse 50, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Svitlana Klishch
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Olha Stilbytska
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Uliana Semaniuk
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
29
|
Chatterjee N, Perrimon N. What fuels the fly: Energy metabolism in Drosophila and its application to the study of obesity and diabetes. SCIENCE ADVANCES 2021; 7:7/24/eabg4336. [PMID: 34108216 PMCID: PMC8189582 DOI: 10.1126/sciadv.abg4336] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/23/2021] [Indexed: 05/16/2023]
Abstract
The organs and metabolic pathways involved in energy metabolism, and the process of ATP production from nutrients, are comparable between humans and Drosophila melanogaster This level of conservation, together with the power of Drosophila genetics, makes the fly a very useful model system to study energy homeostasis. Here, we discuss the major organs involved in energy metabolism in Drosophila and how they metabolize different dietary nutrients to generate adenosine triphosphate. Energy metabolism in these organs is controlled by cell-intrinsic, paracrine, and endocrine signals that are similar between Drosophila and mammals. We describe how these signaling pathways are regulated by several physiological and environmental cues to accommodate tissue-, age-, and environment-specific differences in energy demand. Last, we discuss several genetic and diet-induced fly models of obesity and diabetes that can be leveraged to better understand the molecular basis of these metabolic diseases and thereby promote the development of novel therapies.
Collapse
Affiliation(s)
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
30
|
Hertenstein H, McMullen E, Weiler A, Volkenhoff A, Becker HM, Schirmeier S. Starvation-induced regulation of carbohydrate transport at the blood-brain barrier is TGF-β-signaling dependent. eLife 2021; 10:e62503. [PMID: 34032568 PMCID: PMC8149124 DOI: 10.7554/elife.62503] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
During hunger or malnutrition, animals prioritize alimentation of the brain over other organs to ensure its function and, thus, their survival. This protection, also-called brain sparing, is described from Drosophila to humans. However, little is known about the molecular mechanisms adapting carbohydrate transport. Here, we used Drosophila genetics to unravel the mechanisms operating at the blood-brain barrier (BBB) under nutrient restriction. During starvation, expression of the carbohydrate transporter Tret1-1 is increased to provide more efficient carbohydrate uptake. Two mechanisms are responsible for this increase. Similar to the regulation of mammalian GLUT4, Rab-dependent intracellular shuttling is needed for Tret1-1 integration into the plasma membrane; even though Tret1-1 regulation is independent of insulin signaling. In addition, starvation induces transcriptional upregulation that is controlled by TGF-β signaling. Considering TGF-β-dependent regulation of the glucose transporter GLUT1 in murine chondrocytes, our study reveals an evolutionarily conserved regulatory paradigm adapting the expression of sugar transporters at the BBB.
Collapse
Affiliation(s)
- Helen Hertenstein
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Ellen McMullen
- Institut für Neuro- und Verhaltensbiologie, WWU MünsterMünsterGermany
| | - Astrid Weiler
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Anne Volkenhoff
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| | - Holger M Becker
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
- Division of General Zoology, Department of Biology, University of KaiserslauternKaiserslauternGermany
| | - Stefanie Schirmeier
- Department of Biology, Institute of Zoology, Technische Universität DresdenDresdenGermany
| |
Collapse
|
31
|
Control of Cell Growth and Proliferation by the Tribbles Pseudokinase: Lessons from Drosophila. Cancers (Basel) 2021; 13:cancers13040883. [PMID: 33672471 PMCID: PMC7923445 DOI: 10.3390/cancers13040883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Tribbles pseudokinases represent a sub-branch of the CAMK (Ca2+/calmodulin-dependent protein kinase) subfamily and are associated with disease-associated signaling pathways associated with various cancers, including melanoma, lung, liver, and acute leukemia. The ability of this class of molecules to regulate cell proliferation was first recognized in the model organism Drosophila and the fruit fly genetic model and continues to provide insight into the molecular mechanism by which this family of adapter molecules regulates both normal development and disease associated with corruption of their proper regulation and function. Abstract The Tribbles (Trib) family of pseudokinase proteins regulate cell growth, proliferation, and differentiation during normal development and in response to environmental stress. Mutations in human Trib isoforms (Trib1, 2, and 3) have been associated with metabolic disease and linked to leukemia and the formation of solid tumors, including melanomas, hepatomas, and lung cancers. Drosophila Tribbles (Trbl) was the first identified member of this sub-family of pseudokinases and shares a conserved structure and similar functions to bind and direct the degradation of key mediators of cell growth and proliferation. Common Trib targets include Akt kinase (also known as protein kinase B), C/EBP (CAAT/enhancer binding protein) transcription factors, and Cdc25 phosphatases, leading to the notion that Trib family members stand athwart multiple pathways modulating their growth-promoting activities. Recent work using the Drosophila model has provided important insights into novel facets of conserved Tribbles functions in stem cell quiescence, tissue regeneration, metabolism connected to insulin signaling, and tumor formation linked to the Hippo signaling pathway. Here we highlight some of these recent studies and discuss their implications for understanding the complex roles Tribs play in cancers and disease pathologies.
Collapse
|
32
|
Sodhi K, Denvir J, Liu J, Sanabria JR, Chen Y, Silverstein R, Xie Z, Abraham NG, Shapiro JI. Oxidant-Induced Alterations in the Adipocyte Transcriptome: Role of the Na,K-ATPase Oxidant Amplification Loop. Int J Mol Sci 2020; 21:ijms21165923. [PMID: 32824688 PMCID: PMC7460641 DOI: 10.3390/ijms21165923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/12/2020] [Accepted: 08/15/2020] [Indexed: 12/22/2022] Open
Abstract
(1) Background: Recently we have noted that adipocyte specific expression of the peptide, NaKtide, which was developed to attenuate the Na,K-ATPase oxidant amplification loop, could ameliorate the phenotypical features of uremic cardiomyopathy. We performed this study to better characterize the cellular transcriptomes that are involved in various biological pathways associated with adipocyte function occurring with renal failure. (2) Methods: RNAseq was performed on the visceral adipose tissue of animals subjected to partial nephrectomy. Specific expression of NaKtide in adipocytes was achieved using an adiponectin promoter. To better understand the cause of gene expression changes in vivo, 3T3L1 adipocytes were exposed to indoxyl sulfate (IS) or oxidized low density lipoprotein (oxLDL), with and without pNaKtide (the cell permeant form of NaKtide). RNAseq was also performed on these samples. (3) Results: We noted a large number of adipocyte genes were altered in experimental renal failure. Adipocyte specific NaKtide expression reversed most of these abnormalities. High correlation with some cardiac specific phenotypical features was noted amongst groups of these genes. In the murine adipocytes, both IS and oxLDL induced similar pathway changes as were noted in vivo, and pNaKtide appeared to reverse these changes. Network analysis demonstrated tremendous similarities between the network revealed by gene expression analysis with IS compared with oxLDL, and the combined in vitro dataset was noted to also have considerable similarity to that seen in vivo with experimental renal failure. (4) Conclusions: This study suggests that the myriad of phenotypical features seen with experimental renal failure may be fundamentally linked to oxidant stress within adipocytes.
Collapse
Affiliation(s)
- Komal Sodhi
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (K.S.); (J.D.); (J.L.); (J.R.S.)
| | - James Denvir
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (K.S.); (J.D.); (J.L.); (J.R.S.)
| | - Jiang Liu
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (K.S.); (J.D.); (J.L.); (J.R.S.)
| | - Juan R. Sanabria
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (K.S.); (J.D.); (J.L.); (J.R.S.)
| | - Yiliang Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (Y.C.); (R.S.)
| | - Roy Silverstein
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (Y.C.); (R.S.)
| | - Zijian Xie
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (K.S.); (J.D.); (J.L.); (J.R.S.)
| | - Nader G. Abraham
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, USA;
| | - Joseph I. Shapiro
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (K.S.); (J.D.); (J.L.); (J.R.S.)
- Correspondence: ; Tel.: +1-(304)-691-1704
| |
Collapse
|
33
|
Wang X, Gao H, Xu H. Cluster Analysis of Unhealthy Lifestyles among Elderly Adults with Prediabetes: A Cross-Sectional Study in Rural China. Diabetes Ther 2019; 10:1935-1948. [PMID: 31410710 PMCID: PMC6778567 DOI: 10.1007/s13300-019-00676-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION To explore the clustering and influencing factors of unhealthy lifestyles among elderly adults with prediabetes in rural China. METHODS A cross-sectional study was conducted in Yiyang, Hunan Province, China. Through multistage cluster random sampling and an oral glucose tolerance test, we screened 461 elderly adults aged 60 years and older with prediabetes out of 2144 elderly adults who were sampled. The prediabetic adults completed a survey examining four common lifestyle factors: diet, exercise, smoking, alcohol use and the ability to acquire diabetes-related knowledge and thereby promote one's own health-diabetes health literacy. The influencing factors were analyzed retrospectively with the Kruskal-Wallis test and ordinal logistic regression. RESULTS A total of 425 elderly adults completed the survey, of whom 325 were identified with unhealthy lifestyle clustering. The Kruskal-Wallis test showed significant differences between unhealthy lifestyle clusters by age, gender, marital status, occupation and hyperlipidemia (P < 0.05). Ordinal logistic regression showed that female gender (OR = 0.23, 95% CI: 0.15 0.37), personal annual income ≥ 2800 CNY (OR = 0.61, 95% CI: 0.38 0.99) and occupation as a worker (OR = 0.56, 95% CI: 0.34 0.92) were protective factors against unhealthy lifestyle clustering. An unsatisfactory marital status (OR = 1.60, 95% CI: 1.02 2.51) and low diabetes health literacy (OR = 3.17, 95% CI: 1.03 9.81) were risk factors. CONCLUSION In total, 76.47% of the prediabetic elderly adults in rural China showed unhealthy lifestyle clusters. Being male and having an unsatisfactory marital status, a low personal annual income, an occupation as a farmer and low diabetes health literacy were the main risk factors for unhealthy lifestyle clustering. More effective interventions should be implemented based on these risk factors to prevent diabetes in rural elderly adults.
Collapse
Affiliation(s)
- Xiaofen Wang
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Hailiang Gao
- Department of Human Resources, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huilan Xu
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.
| |
Collapse
|
34
|
Gerlach SU, Sander M, Song S, Herranz H. The miRNA bantam regulates growth and tumorigenesis by repressing the cell cycle regulator tribbles. Life Sci Alliance 2019; 2:2/4/e201900381. [PMID: 31331981 PMCID: PMC6653758 DOI: 10.26508/lsa.201900381] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/15/2022] Open
Abstract
This work identifies the cell cycle regulator tribbles as a target of the miRNA bantam involved in the growth regulatory and oncogenic functions of bantam in Drosophila epithelia. One of the fundamental issues in biology is understanding how organ size is controlled. Tissue growth has to be carefully regulated to generate well-functioning organs, and defects in growth control can result in tumor formation. The Hippo signaling pathway is a universal growth regulator and has been implicated in cancer. In Drosophila, the Hippo pathway acts through the miRNA bantam to regulate cell proliferation and apoptosis. Even though the bantam targets regulating apoptosis have been determined, the target genes controlling proliferation have not been identified thus far. In this study, we identify the gene tribbles as a direct bantam target gene. Tribbles limits cell proliferation by suppressing G2/M transition. We show that tribbles regulation by bantam is central in controlling tissue growth and tumorigenesis. We expand our study to other cell cycle regulators and show that deregulated G2/M transition can collaborate with oncogene activation driving tumor formation.
Collapse
Affiliation(s)
- Stephan U Gerlach
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Moritz Sander
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Shilin Song
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Park YH, Oh EY, Han H, Yang M, Park HJ, Park KH, Lee JH, Park JW. Insulin resistance mediates high-fat diet-induced pulmonary fibrosis and airway hyperresponsiveness through the TGF-β1 pathway. Exp Mol Med 2019; 51:1-12. [PMID: 31133649 PMCID: PMC6536500 DOI: 10.1038/s12276-019-0258-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/31/2018] [Accepted: 01/23/2019] [Indexed: 12/23/2022] Open
Abstract
Prior studies have reported the presence of lung fibrosis and enhanced airway hyperresponsiveness (AHR) in mice with high-fat-diet (HFD)-induced obesity. This study evaluated the role of TGF-β1 in HFD-induced AHR and lung fibrosis in a murine model. We generated HFD-induced obesity mice and performed glucose and insulin tolerance tests. HFD mice with or without ovalbumin sensitization and challenge were also treated with an anti-TGF-β1 neutralizing antibody. AHR to methacholine, inflammatory cells in the bronchoalveolar lavage fluid (BALF), and histological features were evaluated. Insulin was intranasally administered to normal diet (ND) mice, and in vitro insulin stimulation of BEAS-2b cells was performed. HFD-induced obesity mice had increased insulin resistance, enhanced AHR, peribronchial and perivascular fibrosis, and increased numbers of macrophages in the BALF. However, they did not have meaningful eosinophilic or neutrophilic inflammation in the lungs compared with ND mice. The HFD enhanced TGF-β1 expression in the bronchial epithelium, but we found no differences in the expression of interleukin (IL)-4 or IL-5 in lung homogenates. Administration of the anti-TGF-β1 antibody attenuated HFD-induced AHR and lung fibrosis. It also attenuated goblet cell hyperplasia, but did not affect the AHR and inflammatory cell infiltration induced by OVA challenge. The intranasal administration of insulin enhanced TGF-β1 expression in the bronchial epithelium and lung fibrosis. Stimulating BEAS-2b cells with insulin also increased TGF-β1 production by 24 h. We concluded that HFD-induced obesity-associated insulin resistance enhances TGF-β1 expression in the bronchial epithelium, which may play an important role in the development of lung fibrosis and AHR in obesity.
Collapse
Affiliation(s)
- Yoon Hee Park
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Yi Oh
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Heejae Han
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Misuk Yang
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jung Park
- Department of Internal Medicine and Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Hee Park
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Hyun Lee
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Won Park
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea.
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
36
|
Evangelakou Z, Manola M, Gumeni S, Trougakos IP. Nutrigenomics as a tool to study the impact of diet on aging and age-related diseases: the Drosophila approach. GENES & NUTRITION 2019; 14:12. [PMID: 31073342 PMCID: PMC6498619 DOI: 10.1186/s12263-019-0638-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/10/2019] [Indexed: 02/06/2023]
Abstract
Aging is a complex phenomenon caused by the time-dependent loss of cellular homeodynamics and consequently of physiological organismal functions. This process is affected by both genetic and environmental (e.g., diet) factors, as well as by their constant interaction. Consistently, deregulation of nutrient sensing and signaling pathways is considered a hallmark of aging. Nutrigenomics is an emerging scientific discipline that studies changes induced by diet on the genome and thus it considers the intersection of three topics, namely health, diet, and genomics. Model organisms, such as the fruit fly Drosophila melanogaster, have been successfully used for in vivo modeling of higher metazoans aging and for nutrigenomic studies. Drosophila is a well-studied organism with sophisticated genetics and a fully annotated sequenced genome, in which ~ 75% of human disease-related genes have functional orthologs. Also, flies have organs/tissues that perform the equivalent functions of most mammalian organs, while discrete clusters of cells maintain insect carbohydrate homeostasis in a way similar to pancreatic cells. Herein, we discuss the mechanistic connections between nutrition and aging in Drosophila, and how this model organism can be used to study the effect of different diets (including natural products and/or their derivatives) on higher metazoans longevity.
Collapse
Affiliation(s)
- Zoi Evangelakou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Maria Manola
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| |
Collapse
|
37
|
Xu Y, He Z, Song M, Zhou Y, Shen Y. A microRNA switch controls dietary restriction-induced longevity through Wnt signaling. EMBO Rep 2019; 20:embr.201846888. [PMID: 30872315 DOI: 10.15252/embr.201846888] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/31/2019] [Accepted: 02/12/2019] [Indexed: 12/29/2022] Open
Abstract
Dietary restriction (DR) is known to have a potent and conserved longevity effect, yet its underlying molecular mechanisms remain elusive. DR modulates signaling pathways in response to nutrient status, a process that also regulates animal development. Here, we show that the suppression of Wnt signaling, a key pathway controlling development, is required for DR-induced longevity in Caenorhabditis elegans We find that DR induces the expression of mir-235, which inhibits cwn-1/WNT4 expression by binding to the 3'-UTR The "switch-on" of mir-235 by DR occurs at the onset of adulthood, thereby minimizing potential disruptions in development. Our results therefore implicate that DR controls the adult lifespan by using a temporal microRNA switch to modulate Wnt signaling.
Collapse
Affiliation(s)
- Yunpeng Xu
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, Shanghai, China
| | - Zhidong He
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, Shanghai, China
| | - Mengjiao Song
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, Shanghai, China
| | - Yifei Zhou
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, Shanghai, China
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences-University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
38
|
Ávila-Vanzzini N, Michelena HI, Fritche Salazar JF, Herrera-Bello H, Siu Moguel S, Rodríguez Ocampo RR, Oregel Camacho DJ, Espínola Zavaleta N. Clinical and echocardiographic factors associated with mitral plasticity in patients with chronic inferior myocardial infarction. Eur Heart J Cardiovasc Imaging 2019. [PMID: 29529256 DOI: 10.1093/ehjci/jey021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Aims Ischaemic mitral regurgitation (IMR) is consequence of left ventricular (LV) remodelling after myocardial infarction. In some cases, the mitral valve enlarges to compensate for LV remodelling and tenting, improving its coaptation; a process termed 'plasticity'. We sought to identify clinical and echocardiographic factors associated with plasticity in patients with chronic inferior myocardial infarction (CII). Methods and results This study included 91 revascularized CII patients and 46 controls. Plasticity and IMR severity were evaluated by 2D transthoracic echocardiography. Compared with controls, CII patients were older (59 vs. 25 years) and mostly men (80% vs. 46%), both P < 0.001. Chronic inferior myocardial infarction patients also had significant LV remodelling: larger LV volumes, larger mitral tenting areas, larger coaptation depths, longer mitral leaflets and chords, and worse mitral regurgitation (all P ≤ 0.03). Of 91 CII patients, 60 had mitral plasticity (longer anterior and posterior leaflets and longer posterior chords, all P < 0.001), despite not exhibiting significantly larger LV volumes, tenting area or coaptation depth, when compared with patients with no plasticity. Contralateral (anterior) papillary muscle-to-annulus length tended to be increased in CII plasticity patients (P = 0.05). Also they had less moderate and severe IMR (both P < 0.04) compared with non-plasticity CII patients. Multivariate analysis demonstrated independent associations between plasticity and smoking [odds ratio (OR) 0.03, 0.002-0.57; P = 0.019], duration of type-2 diabetes (OR 1.19, 1.007-1.42; P = 0.04) and haemoglobin (OR 2.17, 1.25-3.76; P = 0.005). Conclusion Mitral plasticity results in less moderate and severe IMR. Longer time-duration of diabetes mellitus and higher haemoglobin level are independently associated with mitral plasticity, while smoking independently associates with no plasticity. Increased anterior papillary muscle-to-annulus length in CII patients with plasticity suggests complex LV remodelling mechanisms are involved in plasticity.
Collapse
Affiliation(s)
- Nydia Ávila-Vanzzini
- Department of Out patients Care, Echocardiography, Nuclear medicine National Instituto of Cardiology Ignacio Chávez, Juan Badiano No.1, Colonia Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Hector I Michelena
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St Sw, Rochester, MN, USA
| | - Juan Francisco Fritche Salazar
- Department of Out patients Care, Echocardiography, Nuclear medicine National Instituto of Cardiology Ignacio Chávez, Juan Badiano No.1, Colonia Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Héctor Herrera-Bello
- Intermediate Care Unit Medica Sur Clinical Foundation, Puente de Piedra 150, Toriello Guerra, Delegación Tlalpan, Ciudad de México, Mexico City, Mexico
| | - Silvia Siu Moguel
- Hospital Regional ISSSTE, Av Díaz Mirón SN Colonia, Moderno, 91910 Veracruz, Mexico
| | - Rubén Rafael Rodríguez Ocampo
- Autonomous University of Nayarit, Edificio de la Unidad Academica de Medicina, Ciudad de la cultura "Amado Nervo" CP: 63000 Tepic Nayarit, Mexico
| | - Diego Javier Oregel Camacho
- Autonomous University of Nayarit, Edificio de la Unidad Academica de Medicina, Ciudad de la cultura "Amado Nervo" CP: 63000 Tepic Nayarit, Mexico
| | - Nilda Espínola Zavaleta
- Department of Out patients Care, Echocardiography, Nuclear medicine National Instituto of Cardiology Ignacio Chávez, Juan Badiano No.1, Colonia Sección XVI, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
39
|
Abstract
The insect fat body is analogous to vertebrate adipose tissue and liver. In this review, the new and exciting advancements made in fat body biology in the last decade are summarized. Controlled by hormonal and nutritional signals, insect fat body cells undergo mitosis during embryogenesis, endoreplication during the larval stages, and remodeling during metamorphosis and regulate reproduction in adults. Fat body tissues are major sites for nutrient storage, energy metabolism, innate immunity, and detoxification. Recent studies have revealed that the fat body plays a central role in the integration of hormonal and nutritional signals to regulate larval growth, body size, circadian clock, pupal diapause, longevity, feeding behavior, and courtship behavior, partially by releasing fat body signals to remotely control the brain. In addition, the fat body has emerged as a fascinating model for studying metabolic disorders and immune diseases. Potential future directions for fat body biology are also proposed herein.
Collapse
Affiliation(s)
- Sheng Li
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, Guangdong 510631, China; , ,
| | - Xiaoqiang Yu
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, Guangdong 510631, China; , ,
| | - Qili Feng
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, Guangdong 510631, China; , ,
| |
Collapse
|
40
|
Using Mouse and Drosophila Models to Investigate the Mechanistic Links between Diet, Obesity, Type II Diabetes, and Cancer. Int J Mol Sci 2018; 19:ijms19124110. [PMID: 30567377 PMCID: PMC6320797 DOI: 10.3390/ijms19124110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023] Open
Abstract
Many of the links between diet and cancer are controversial and over simplified. To date, human epidemiological studies consistently reveal that patients who suffer diet-related obesity and/or type II diabetes have an increased risk of cancer, suffer more aggressive cancers, and respond poorly to current therapies. However, the underlying molecular mechanisms that increase cancer risk and decrease the response to cancer therapies in these patients remain largely unknown. Here, we review studies in mouse cancer models in which either dietary or genetic manipulation has been used to model obesity and/or type II diabetes. These studies demonstrate an emerging role for the conserved insulin and insulin-like growth factor signaling pathways as links between diet and cancer progression. However, these models are time consuming to develop and expensive to maintain. As the world faces an epidemic of obesity and type II diabetes we argue that the development of novel animal models is urgently required. We make the case for Drosophila as providing an unparalleled opportunity to combine dietary manipulation with models of human metabolic disease and cancer. Thus, combining diet and cancer models in Drosophila can rapidly and significantly advance our understanding of the conserved molecular mechanisms that link diet and diet-related metabolic disorders to poor cancer patient prognosis.
Collapse
|
41
|
Bayliak MM, Abrat OB, Storey JM, Storey KB, Lushchak VI. Interplay between diet-induced obesity and oxidative stress: Comparison between Drosophila and mammals. Comp Biochem Physiol A Mol Integr Physiol 2018; 228:18-28. [PMID: 30385171 DOI: 10.1016/j.cbpa.2018.09.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022]
Abstract
Obesity caused by excessive fat accumulation in adipocytes is a growing global problem and is a major contributing risk factor for many chronic metabolic diseases. There is increasing evidence that oxidative stress plays a crucial role in both obesity progression and obesity-related complications. In recent years, Drosophila models of diet-induced obesity and associated pathologies have been successfully developed through manipulation of carbohydrate or fat concentrations in the food. Obese flies accumulate triacylglycerols in the fat body, an organ homologous to mammalian adipose tissue and exhibit metabolic and physiological complications including hyperglycemia, redox imbalance and shortened longevity; these are all similar to those observed in obese humans. In this review, we summarize current data on the mechanisms of oxidative stress induction in obesity, with emphasis on metabolic switches and the involvement of redox-responsive signaling pathways such as NF-κB and Nfr2. The recent achievements with D. melanogaster model suggest a complicated relationship between obesity, oxidative stress, and longevity but the Drosophila model offers probably the best opportunities to delve further into unraveling these interactions, particularly the roles of antioxidants and of Nrf2-regulated responses, in order to increase our understanding of the obese metabolic phenotype and test and develop anti-obesity pharmaceuticals.
Collapse
Affiliation(s)
- Maria M Bayliak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk 76018, Ukraine.
| | - Olexandra B Abrat
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk 76018, Ukraine.
| | - Janet M Storey
- 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.
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str, Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
42
|
Gáliková M, Klepsatel P. Obesity and Aging in the Drosophila Model. Int J Mol Sci 2018; 19:ijms19071896. [PMID: 29954158 PMCID: PMC6073435 DOI: 10.3390/ijms19071896] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Being overweight increases the risk of many metabolic disorders, but how it affects lifespan is not completely clear. Not all obese people become ill, and the exact mechanism that turns excessive fat storage into a health-threatening state remains unknown. Drosophila melanogaster has served as an excellent model for many diseases, including obesity, diabetes, and hyperglycemia-associated disorders, such as cardiomyopathy or nephropathy. Here, we review the connections between fat storage and aging in different types of fly obesity. Whereas obesity induced by high-fat or high-sugar diet is associated with hyperglycemia, cardiomyopathy, and in some cases, shortening of lifespan, there are also examples in which obesity correlates with longevity. Transgenic lines with downregulations of the insulin/insulin-like growth factor (IIS) and target of rapamycin (TOR) signaling pathways, flies reared under dietary restriction, and even certain longevity selection lines are obese, yet long-lived. The mechanisms that underlie the differential lifespans in distinct types of obesity remain to be elucidated, but fat turnover, inflammatory pathways, and dysregulations of glucose metabolism may play key roles. Altogether, Drosophila is an excellent model to study the physiology of adiposity in both health and disease.
Collapse
Affiliation(s)
- Martina Gáliková
- Department of Zoology, Stockholm University, Svante Arrhenius väg 18B, S-106 91 Stockholm, Sweden.
| | - Peter Klepsatel
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 06 Bratislava, Slovakia.
| |
Collapse
|
43
|
de Paula MT, Silva MRP, Araújo SM, Bortolotto VC, Martins IK, Macedo GE, Franco JL, Posser T, Prigol M. Drosophila melanogaster: A model to study obesity effects on genes expression and developmental changes on descendants. J Cell Biochem 2018; 119:5551-5562. [PMID: 29377237 DOI: 10.1002/jcb.26724] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022]
Abstract
Maternal obesity and metabolic diseases are two of the most important potential dangers to offspring, given that impaired offspring may cause deficiencies that impair the adult life and health. This study evaluated the oxidative damage, the enzymatic antioxidant defenses, and the enzymes of fatty acid metabolism, such as Acyl-CoA Synthetase and Acetyl-CoA Synthetase (mRNA expression levels), as well as the modulation of cell stress signaling pathway, as Hsp83, and gene expression and insulin-like peptide DILP6 in Drosophila melanogaster models that received a high fat diet (HFD) (10% and 20% of coconut oil) throughout their development period. After 7 days, the progenitor flies were removed and, the remaining eggs were monitored daily, until the eclosion. The descendants were then exposed to a regular diet (RD). The results revealed that the HFD caused a decrease in the proportion of eclosion, lifespan, MTT reduction in mitochondrial enriched fractions, AceCS1 levels, mRNA expression levels (SOD and CAT), and in catalase activity a decrease was only observed in the group that received the highest concentration of coconut oil. In parallel, it was demonstrated an increase in the upregulation of HSP83 mRNA levels, but only when 10% of coconut oil was added, and an increase in glucose and triglyceride levels, as well as in DILP6 mRNA levels in larger concentration of coconut oil tested (20%). In conclusion, flies that have progenitors fed with HFD can develop metabolic dysfunctions, causing oxidative insults, which are involved in the shortening of lifespan.
Collapse
Affiliation(s)
- Mariane T de Paula
- Laboratório de Avaliações Farmacológicas e Toxicológicas aplicadas às Moléculas Bioativas-Unipampa, Universidade Federal do Pampa, Itaqui, RS, Brazil
| | - Márcia R P Silva
- Laboratório de Avaliações Farmacológicas e Toxicológicas aplicadas às Moléculas Bioativas-Unipampa, Universidade Federal do Pampa, Itaqui, RS, Brazil
| | - Stífani M Araújo
- Laboratório de Avaliações Farmacológicas e Toxicológicas aplicadas às Moléculas Bioativas-Unipampa, Universidade Federal do Pampa, Itaqui, RS, Brazil
| | - Vandreza C Bortolotto
- Laboratório de Avaliações Farmacológicas e Toxicológicas aplicadas às Moléculas Bioativas-Unipampa, Universidade Federal do Pampa, Itaqui, RS, Brazil
| | - Illana K Martins
- Centro Interdisciplinar de Pesquisa em Biotecnologia (CIP/BIOTEC), Universidade Federal do Pampa, São Gabriel, RS, Brazil
| | - Giulianna E Macedo
- Centro Interdisciplinar de Pesquisa em Biotecnologia (CIP/BIOTEC), Universidade Federal do Pampa, São Gabriel, RS, Brazil
| | - Jeferson L Franco
- Centro Interdisciplinar de Pesquisa em Biotecnologia (CIP/BIOTEC), Universidade Federal do Pampa, São Gabriel, RS, Brazil
| | - Thaís Posser
- Centro Interdisciplinar de Pesquisa em Biotecnologia (CIP/BIOTEC), Universidade Federal do Pampa, São Gabriel, RS, Brazil
| | - Marina Prigol
- Laboratório de Avaliações Farmacológicas e Toxicológicas aplicadas às Moléculas Bioativas-Unipampa, Universidade Federal do Pampa, Itaqui, RS, Brazil
| |
Collapse
|
44
|
Abstract
Excess adipose fat accumulation, or obesity, is a growing problem worldwide in terms of both the rate of incidence and the severity of obesity-associated metabolic disease. Adipose tissue evolved in animals as a specialized dynamic lipid storage depot: adipose cells synthesize fat (a process called lipogenesis) when energy is plentiful and mobilize stored fat (a process called lipolysis) when energy is needed. When a disruption of lipid homeostasis favors increased fat synthesis and storage with little turnover owing to genetic predisposition, overnutrition or sedentary living, complications such as diabetes and cardiovascular disease are more likely to arise. The vinegar fly Drosophila melanogaster (Diptera: Drosophilidae) is used as a model to better understand the mechanisms governing fat metabolism and distribution. Flies offer a wealth of paradigms with which to study the regulation and physiological effects of fat accumulation. Obese flies accumulate triacylglycerols in the fat body, an organ similar to mammalian adipose tissue, which specializes in lipid storage and catabolism. Discoveries in Drosophila have ranged from endocrine hormones that control obesity to subcellular mechanisms that regulate lipogenesis and lipolysis, many of which are evolutionarily conserved. Furthermore, obese flies exhibit pathophysiological complications, including hyperglycemia, reduced longevity and cardiovascular function - similar to those observed in obese humans. Here, we review some of the salient features of the fly that enable researchers to study the contributions of feeding, absorption, distribution and the metabolism of lipids to systemic physiology.
Collapse
Affiliation(s)
- Laura Palanker Musselman
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY 13902, USA
| | - Ronald P Kühnlein
- Department of Biochemistry 1, Institute of Molecular Biosciences, University of Graz, Humboldtstraβe 50/II, A-8010 Graz, Austria.,BioTechMed-Graz, Graz, Austria
| |
Collapse
|
45
|
Herranz H, Cohen SM. Drosophila as a Model to Study the Link between Metabolism and Cancer. J Dev Biol 2017; 5:E15. [PMID: 29615570 PMCID: PMC5831792 DOI: 10.3390/jdb5040015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022] Open
Abstract
Cellular metabolism has recently been recognized as a hallmark of cancer. Investigating the origin and effects of the reprogrammed metabolism of tumor cells, and identifying its genetic mediators, will improve our understanding of how these changes contribute to disease progression and may suggest new approaches to therapy. Drosophila melanogaster is emerging as a valuable model to study multiple aspects of tumor formation and malignant transformation. In this review, we discuss the use of Drosophila as model to study how changes in cellular metabolism, as well as metabolic disease, contribute to cancer.
Collapse
Affiliation(s)
- Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200 N Copenhagen, Denmark.
| | - Stephen M Cohen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200 N Copenhagen, Denmark.
| |
Collapse
|
46
|
Galeone A, Han SY, Huang C, Hosomi A, Suzuki T, Jafar-Nejad H. Tissue-specific regulation of BMP signaling by Drosophila N-glycanase 1. eLife 2017; 6:27612. [PMID: 28826503 PMCID: PMC5599231 DOI: 10.7554/elife.27612] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 08/03/2017] [Indexed: 12/14/2022] Open
Abstract
Mutations in the human N-glycanase 1 (NGLY1) cause a rare, multisystem congenital disorder with global developmental delay. However, the mechanisms by which NGLY1 and its homologs regulate embryonic development are not known. Here we show that Drosophila Pngl encodes an N-glycanase and exhibits a high degree of functional conservation with human NGLY1. Loss of Pngl results in developmental midgut defects reminiscent of midgut-specific loss of BMP signaling. Pngl mutant larvae also exhibit a severe midgut clearance defect, which cannot be fully explained by impaired BMP signaling. Genetic experiments indicate that Pngl is primarily required in the mesoderm during Drosophila development. Loss of Pngl results in a severe decrease in the level of Dpp homodimers and abolishes BMP autoregulation in the visceral mesoderm mediated by Dpp and Tkv homodimers. Thus, our studies uncover a novel mechanism for the tissue-specific regulation of an evolutionarily conserved signaling pathway by an N-glycanase enzyme. DNA carries the information needed to build and maintain an organism, and units of DNA known as genes contain coded instructions to build other molecules, including enzymes. Sometimes, genes can become faulty and develop mutations that can affect how an embryo develops and lead to diseases. For example, people with mutations in the gene that encodes an enzyme called N-glycanase 1 experience many problems with their nervous system, gut and other organs. Normally, N-glycanase 1 helps the body remove specific sugar molecules from some proteins in the cells, and is also thought to be important during embryonic development. As an embryo develops, its cells undergo a series of transformations, which is regulated by different molecules and signaling pathways. For example, a pathway known as BMP signaling plays an important role in many tissues. Problems with this pathway can lead to many diseases throughout the body, including the gut, where it helps cells to develop. Previous research has shown that fruit flies lacking the gene that codes for an equivalent N-glycanase enzyme (which is called Pngl in flies) cannot develop properly into adults. However, until now it was not known what type of cells need the N-glycanase enzyme in any organism, or if NGLY1 is essential for important signaling pathways like BMP signaling. Now, Galeone et al. have used genetically modified flies to test how losing Pngl affected their development. The results first showed that engineering Pngl-deficient fruit flies to produce the human enzyme eliminated their problems; these flies developed and survived like normal flies. This confirmed that that the human and fly enzymes can perform equivalent roles. Galeone et al. then discovered that Pngl plays two distinct roles in a group of cells that surround the fruit fly’s gut tissue and give rise to the cells that eventually form the muscle layer in the gut. In the larvae, Pngl was required to empty the gut, which is a necessary step before the larvae can develop into an adult. Moreover, Pngl is needed for BMP signaling in the gut, and when flies had the enzyme removed, some parts of their gut could not from properly. This study will provide a framework to improve our understanding of how BMP signaling is regulated in humans. A next step will be to test if some of the symptoms experienced by patients without a working copy of the gene for N-glycanase 1 are caused by a faulty BMP-signaling system in specific tissues. If this is the case, it could provide new opportunities to treat some of these symptoms.
Collapse
Affiliation(s)
- Antonio Galeone
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Seung Yeop Han
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Chengcheng Huang
- Glycometabolome Team, RIKEN Global Research Cluster, Saitama, Japan
| | - Akira Hosomi
- Glycometabolome Team, RIKEN Global Research Cluster, Saitama, Japan
| | - Tadashi Suzuki
- Glycometabolome Team, RIKEN Global Research Cluster, Saitama, Japan
| | - Hamed Jafar-Nejad
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| |
Collapse
|