1
|
Li H, Zhang J, Fu Y, Zhang Y, Zhang C, Sun X, Wu F, He J. Antidiabetic compounds 8a, 8b, 8k, and 9h enhance insulin secretion: activity and mechanism. Endocrine 2021; 71:365-377. [PMID: 33219494 DOI: 10.1007/s12020-020-02537-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE This study primarily investigated the effects of hypoglycemic compounds (Imeglimin derivatives) on insulin secretion in type 2 diabetes mellitus (T2DM), and further explored the possible mechanism underlying these effects. METHODS Firstly, Metformin was used as the initiating compound to synthesize three sets of derivatives which contained Imeglimin structure core. At the cellular level, we screened compounds with better effect on the activity of insulin receptor tyrosine protein kinase (IFcTPK) after the islet β cells were treated with the compounds of different concentrations. The insulin secretion was assessed using radioimmunoassay and the cytotoxicity to islet β cells was evaluated by means of MTT assay following treatment with the compounds. The Ca2+-related mechanism by which these compounds promote insulin secretion was elucidated with whole cell recordings from current-clamp mode. RESULTS Totally, 48 synthesized compounds were generated, wherein 10 compounds could increase the activity of IFcTPK in HIT-T15 cells better among these compounds. The modified Imeglimin, especially in the structure of hydrophilic hydroxyl or piperidine rings, could improve the activity of the compound to promote insulin secretion. Furthermore, the compounds 8a, 8b, 8k, and 9h revealed high insulin secretion-promoting activity. These compounds enhanced insulin secretion in islet β cells by repressing the ATP-sensitive K(+) and voltage-gated K+ pathway. CONCLUSIONS Our findings indicate that the hypoglycemic compounds 8a, 8b, 8k, and 9h confer better promotive effect on insulin secretion, which provides a reference for the development of drugs with better hypoglycemic activity.
Collapse
Affiliation(s)
- Hui Li
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Jian Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Yongli Fu
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Yixin Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Chunhui Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Xiaozhu Sun
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China
| | - Fang Wu
- Department of Hepatology, The Seventh Hospital of Qiqihar, Qiqihar, 161000, PR China
| | - Jing He
- Department of Endocrinology, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161000, PR China.
| |
Collapse
|
2
|
Xu DQ, Li CJ, Jiang ZZ, Wang L, Huang HF, Li ZJ, Sun LX, Fan SS, Zhang LY, Wang T. The hypoglycemic mechanism of catalpol involves increased AMPK-mediated mitochondrial biogenesis. Acta Pharmacol Sin 2020; 41:791-799. [PMID: 31937931 PMCID: PMC7470840 DOI: 10.1038/s41401-019-0345-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022] Open
Abstract
Mitochondria serve as sensors of energy regulation and glucose levels, which are impaired by diabetes progression. Catalpol is an iridoid glycoside that exerts a hypoglycemic effect by improving mitochondrial function, but the underlying mechanism has not been fully elucidated. In the current study we explored the effects of catalpol on mitochondrial function in db/db mice and C2C12 myotubes in vitro. After oral administration of catalpol (200 mg·kg−1·d−1) for 8 weeks, db/db mice exhibited a decreased fasting blood glucose level and restored mitochondrial function in skeletal muscle. Catalpol increased mitochondrial biogenesis, evidenced by significant elevations in the number of mitochondria, mitochondrial DNA levels, and the expression of three genes associated with mitochondrial biogenesis: peroxisome proliferator-activated receptor gammaco-activator 1 (PGC-1α), mitochondrial transcription factor A (TFAM) and nuclear respiratory factor 1 (NRF1). In C2C12 myotubes, catalpol significantly increased glucose uptake and ATP production. These effects depended on activation of AMP-activated protein kinase (AMPK)-mediated mitochondrial biogenesis. Thus, catalpol improves skeletal muscle mitochondrial function by activating AMPK-mediated mitochondrial biogenesis. These findings may guide the development of a new therapeutic approach for type 2 diabetes.
Collapse
|
3
|
Selleri L, Zappavigna V, Ferretti E. 'Building a perfect body': control of vertebrate organogenesis by PBX-dependent regulatory networks. Genes Dev 2019; 33:258-275. [PMID: 30824532 PMCID: PMC6411007 DOI: 10.1101/gad.318774.118] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pbx genes encode transcription factors that belong to the TALE (three-amino-acid loop extension) superclass of homeodomain proteins. We have witnessed a surge in information about the roles of this gene family as leading actors in the transcriptional control of development. PBX proteins represent a clear example of how transcription factors can regulate developmental processes by combinatorial properties, acting within multimeric complexes to implement activation or repression of transcription depending on their interaction partners. Here, we revisit long-emphasized functions of PBX transcription factors as cofactors for HOX proteins, major architects of the body plan. We further discuss new knowledge on roles of PBX proteins in different developmental contexts as upstream regulators of Hox genes-as factors that interact with non-HOX proteins and can work independently of HOX-as well as potential pioneer factors. Committed to building a perfect body, PBX proteins govern regulatory networks that direct essential morphogenetic processes and organogenesis in vertebrate development. Perturbations of PBX-dependent networks can cause human congenital disease and cancer.
Collapse
Affiliation(s)
- Licia Selleri
- Program in Craniofacial Biology, University of California at San Francisco, San Francisco, California 94143, USA
- Institute of Human Genetics, University of California at San Francisco, San Francisco, California 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Orofacial Sciences, University of California at San Francisco, San Francisco, California 94143, USA
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| | - Vincenzo Zappavigna
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Elisabetta Ferretti
- The Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
4
|
Yu P, Xu X, Zhang J, Xia X, Xu F, Weng J, Lai X, Shen Y. Liraglutide Attenuates Nonalcoholic Fatty Liver Disease through Adjusting Lipid Metabolism via SHP1/AMPK Signaling Pathway. Int J Endocrinol 2019; 2019:1567095. [PMID: 31236111 PMCID: PMC6545813 DOI: 10.1155/2019/1567095] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
A glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide (LR) had been experimentally and clinically shown to ameliorate nonalcoholic fatty liver disease (NAFLD). This study aimed to investigate the beneficial effect of LR on NAFLD in vivo and in vitro and its underlying molecular mechanism. The effects of LR were examined on the high-fat diet-induced in vivo model in mice and in vitro model of NAFLD in human HepG2 cells. Liver tissues and HepG2 cells were procured for measuring lipid metabolism, histological examination, and western blot analysis. LR administration significantly lowered the serum lipid profile and lipid disposition in vitro and in vivo because of the altered expression of enzymes on hepatic gluconeogenesis and lipid metabolism. Moreover, LR significantly decreased Src homology region 2 domain-containing phosphatase-1 (SHP1) and then increased the expression of phosphorylated-AMP-activated protein kinase (p-AMPK). However, the overexpression of SHP1 mediated by lentivirus vector reversed LR-induced improvement in lipid deposition. Moreover, SHP1 silencing could further increase the expression of p-AMPK to ameliorate lipid metabolism and relative lipogenic gene induced by LR. In addition, abrogation of AMPK by Compound C eliminated the protective effects of LR on lipid metabolism without changing the expression of SHP1. LR markedly prevented NAFLD through adjusting lipid metabolism via SHP1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, Jiangxi Institute of Endocrine and Metabolic Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xi Xu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Xia
- Department of Physiology and Pathophysiology, College of Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Xiaoyang Lai
- Department of Endocrinology and Metabolism, Jiangxi Institute of Endocrine and Metabolic Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, Jiangxi Institute of Endocrine and Metabolic Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Ye D, Lou G, Zhang T, Dong F, Liu Y. MiR-17 family-mediated regulation of Pknox1 influences hepatic steatosis and insulin signaling. J Cell Mol Med 2018; 22:6167-6175. [PMID: 30338914 PMCID: PMC6237553 DOI: 10.1111/jcmm.13902] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
The aberrant expression of Pknox1 is associated with hepatic glucose and lipid dysmetabolism status of type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD). However, the underlying mechanism causing Pknox1 overexpression in this pathological status remains unclear. By using miRNA target prediction programs, we found that the 3′‐UTR of the Pknox1 mRNA sequence contains highly conserved target sites of miR‐17 family. In a rat model of streptozotocin and high‐fat diet‐induced T2DM and NAFLD complication, the increased hepatic expression of Pknox1 was consistent with decreased expressions of miR‐17 family, especially miR‐17 and miR‐20a. Furthermore, an inverse correlation was observed between Pknox1 and miR‐17 and miR‐20a in free fatty acids‐induced hepatocyte steatosis. Dual‐luciferase reporter assay further showed that Pknox1 was a valid target gene of miR‐17 family. The ectopic expression of miR‐17 or miR‐20a could markedly suppress Pknox1 expression in hepatocytes. MiR‐17 or miR‐20a overexpression also resulted in significantly enhanced insulin sensitivity and reduced hepatocyte steatosis in HepG2 and L02 cells, which were determined by altered phosphorylation on insulin receptor signaling pathway proteins and decreased intracellular triglyceride and lipid accumulation, respectively. These data implicate the upregulated hepatic expression of Pknox1 in T2DM complicated with NAFLD may be caused by the reduced expression of miR‐17 family, indicating that developing miRNA‐mediated regulation strategies on Pknox1 may provide new therapeutic options for metabolic disease.
Collapse
Affiliation(s)
- Dan Ye
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Guohua Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatament of Infectious Diseases, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Tianbao Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatament of Infectious Diseases, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Fengqin Dong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatament of Infectious Diseases, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Moreau P, Cournac A, Palumbo GA, Marbouty M, Mortaza S, Thierry A, Cairo S, Lavigne M, Koszul R, Neuveut C. Tridimensional infiltration of DNA viruses into the host genome shows preferential contact with active chromatin. Nat Commun 2018; 9:4268. [PMID: 30323189 PMCID: PMC6189100 DOI: 10.1038/s41467-018-06739-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/19/2018] [Indexed: 01/05/2023] Open
Abstract
Whether non-integrated viral DNAs distribute randomly or target specific positions within the higher-order architecture of mammalian genomes remains largely unknown. Here we use Hi-C and viral DNA capture (CHi-C) in primary human hepatocytes infected by either hepatitis B virus (HBV) or adenovirus type 5 (Ad5) virus to show that they adopt different strategies in their respective positioning at active chromatin. HBV contacts preferentially CpG islands (CGIs) enriched in Cfp1 a factor required for its transcription. These CGIs are often associated with highly expressed genes (HEG) and genes deregulated during infection. Ad5 DNA interacts preferentially with transcription start sites (TSSs) and enhancers of HEG, as well as genes upregulated during infection. These results show that DNA viruses use different strategies to infiltrate genomic 3D networks and target specific regions. This targeting may facilitate the recruitment of transcription factors necessary for their own replication and contribute to the deregulation of cellular gene expression.
Collapse
Affiliation(s)
- Pierrick Moreau
- Institut Pasteur, Unité Hepacivirus et Immunité Innée, 75015, Paris, France.,CNRS, UMR 3569, 75015, Paris, France.,Institut Pasteur, Département de Virologie, Paris, France
| | - Axel Cournac
- Institut Pasteur, Département Génomes et Génétique, Groupe Régulation spatiale des génomes, 75015, Paris, France.,CNRS, UMR 3525, 75015, Paris, France
| | - Gianna Aurora Palumbo
- Institut Pasteur, Unité Hepacivirus et Immunité Innée, 75015, Paris, France.,CNRS, UMR 3569, 75015, Paris, France.,Institut Pasteur, Département de Virologie, Paris, France
| | - Martial Marbouty
- Institut Pasteur, Département Génomes et Génétique, Groupe Régulation spatiale des génomes, 75015, Paris, France.,CNRS, UMR 3525, 75015, Paris, France
| | - Shogofa Mortaza
- Institut Pasteur, Département Génomes et Génétique, Groupe Régulation spatiale des génomes, 75015, Paris, France.,CNRS, UMR 3525, 75015, Paris, France
| | - Agnes Thierry
- Institut Pasteur, Département Génomes et Génétique, Groupe Régulation spatiale des génomes, 75015, Paris, France.,CNRS, UMR 3525, 75015, Paris, France
| | - Stefano Cairo
- XenTech, Research and Development Department, 91000, Evry, France
| | - Marc Lavigne
- Institut Pasteur, Département de Virologie, Paris, France.,Institut Cochin-INSERM U1016-CNRS UMR8104, Université Paris Descartes, Paris, France
| | - Romain Koszul
- Institut Pasteur, Département Génomes et Génétique, Groupe Régulation spatiale des génomes, 75015, Paris, France. .,CNRS, UMR 3525, 75015, Paris, France.
| | - Christine Neuveut
- Institut Pasteur, Unité Hepacivirus et Immunité Innée, 75015, Paris, France. .,CNRS, UMR 3569, 75015, Paris, France. .,Institut Pasteur, Département de Virologie, Paris, France.
| |
Collapse
|
7
|
Liotti A, Cabaro S, Cimmino I, Ricci S, Procaccini C, Paciello O, Raciti GA, Spinelli R, Iossa S, Matarese G, Miele C, Formisano P, Beguinot F, Oriente F. Prep1 deficiency improves metabolic response in white adipose tissue. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:515-525. [PMID: 29474930 DOI: 10.1016/j.bbalip.2018.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 02/09/2018] [Accepted: 02/17/2018] [Indexed: 12/13/2022]
Abstract
Prep1 is a gene encoding for a homeodomain transcription factor which induces hepatic and muscular insulin resistance. In this study, we show that Prep1 hypomorphic heterozygous (Prep1i/+) mice, expressing low levels of protein, featured a 23% and a 25% reduction of total body lipid content and epididymal fat, respectively. The percentage of the small adipocytes (25-75 μm) was 30% higher in Prep1i/+ animals than in the WT, with a reciprocal difference in the large adipose cells (100-150 and >150 μm). Insulin-stimulated insulin receptor tyrosine and Akt serine phosphorylation markedly increased in Prep1i/+ mice, paralleled by 3-fold higher glucose uptake and a significant increase of proadipogenic genes such as C/EBPα, GLUT4, and FABP4. Moreover, T cells infiltration and TNF-α, IFNγ and leptin expression were reduced in adipose tissue from Prep1i/+ mice, while adiponectin levels were 2-fold higher. Furthermore, Prep1i/+ mature adipocytes released lower amounts of pro-inflammatory cytokines and higher amount of adiponectin compared to WT cells. Incubation of murine liver cell line (NMuLi) with conditioned media (CM) from mature adipocytes of Prep1i/+ mice improved glucose metabolism, while those from WT mice had no effect. Consistent with these data, Prep1 overexpression in 3T3-L1 adipocytes impaired adipogenesis and insulin signaling, and increased proinflammatory cytokine secretion. All these findings suggest that Prep1 silencing reduces inflammatory response and increases insulin sensitivity in adipose tissue. In addition, CM from mature adipocytes of Prep1i/+ mice improve metabolism in hepatic cells.
Collapse
Affiliation(s)
- Antonietta Liotti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Ricci
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Claudio Procaccini
- Laboratory of Immunology, National Council of Research (CNR), Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, Federico II University of Naples, Naples, Italy
| | - Gregory A Raciti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Rosa Spinelli
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Susanna Iossa
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Giuseppe Matarese
- Laboratory of Immunology, National Council of Research (CNR), Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy.
| |
Collapse
|
8
|
Prep1 Deficiency Affects Olfactory Perception and Feeding Behavior by Impairing BDNF-TrkB Mediated Neurotrophic Signaling. Mol Neurobiol 2018; 55:6801-6815. [PMID: 29349576 PMCID: PMC6061220 DOI: 10.1007/s12035-018-0873-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/07/2018] [Indexed: 11/01/2022]
Abstract
Prep1 is a homeodomain transcription factor which has an important role in hindbrain development. Prep1 expression is also kept in adult mouse brain and in particular within the olfactory bulbs. Moreover, many Prep1 neurons co-localize with Calbindin-positive periglomerular interneurons in olfactory glomerular layer. However, Prep1 function in this brain region is still unknown. In this study, we show that Prep1 hypomorphic heterozygous (Prep1i/+) mice express low levels of protein and feature a 30% reduction of olfactory bulb area, compared to WT mice. In addition, Prep1i/+ mice olfactory bulb histological analysis indicated a 20% lower cytochrome C oxidase activity within the glomerular layer, accompanied by a reduced number of periglomerular interneurons, compared to the WT littermates. Consistently, olfactory perception test highlighted that Prep1 hypomorphic heterozygous mice display a scant ability to distinguish odors, which significantly impacts on feeding behavior, as Prep1i/+ mice revealed a reduced preference for high-fat food. Analysis of BDNF signaling, which represents the main molecular mediator of olfactory plasticity, showed that Prep1i/+ mouse olfactory bulbs feature a 30% reduction of TrkB receptor levels and a decreased activation of ERK1/2. Similarly, overexpression of Prep1 in mouse neuronal cells (N2A) caused an increase of TrkB expression levels, BDNF-induced ERK phosphorylation, and cell viability, compared to control cells. We conclude that Prep1 deficiency alters olfactory morpho-functional integrity and olfaction-mediated eating behavior by affecting BDNF-TrkB signaling. Prep1 could, therefore, play a crucial role in behavioral dysfunctions associated to impaired responsiveness to BDNF.
Collapse
|
9
|
Oriente F, Perruolo G, Cimmino I, Cabaro S, Liotti A, Longo M, Miele C, Formisano P, Beguinot F. Prep1, A Homeodomain Transcription Factor Involved in Glucose and Lipid Metabolism. Front Endocrinol (Lausanne) 2018; 9:346. [PMID: 30002646 PMCID: PMC6032887 DOI: 10.3389/fendo.2018.00346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/11/2018] [Indexed: 12/28/2022] Open
Abstract
The three-amino acid loop extension (TALE) homeodomain proteins are a family of transcription factor including the mammalian Pbx, MEIS and Prep proteins. TALE proteins can bind other transcription factors such as Pdx-1 and play an important role in the regulation of glucose metabolism. Experiments performed in mutant mice have shown that while the single Pbx1 or Pdx-1 knockout mice feature pancreatic islet malformations, impaired glucose tolerance and hypoinsulinemia, the trans-heterozygous Pbx1+/-Pdx1+/- mice develop age-dependent overt diabetes mellitus. In contrast, Prep1 plays a different role with respect to these proteins. Indeed, Prep1 hypomorphic mice, expressing low levels of protein, feature pancreatic islet hypoplasia accompanied by hypoinsulinemia similar to Pbx1 or Pdx1. Nevertheless, these animals show increased insulin sensitivity in skeletal muscle, liver and adipose tissue accompanied by protection from streptozotocin-induced diabetes. In addition, Prep1 hypomorphic mice feature reduced triglyceride synthesis and do not develop steatohepatitis after a methionine and coline deficient diet. In this review we have underlined how important metabolic functions are controlled by TALE proteins, in particular by Prep1, leading to hypothesis that its suppression might represent beneficial effect in the care of metabolic diseases.
Collapse
Affiliation(s)
- Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Giuseppe Perruolo
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Antonietta Liotti
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Michele Longo
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
- *Correspondence: Pietro Formisano
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
10
|
Cimmino I, Lorenzo V, Fiory F, Doti N, Ricci S, Cabaro S, Liotti A, Vitagliano L, Longo M, Miele C, Formisano P, Beguinot F, Ruvo M, Oriente F. A peptide antagonist of Prep1-p160 interaction improves ceramide-induced insulin resistance in skeletal muscle cells. Oncotarget 2017; 8:71845-71858. [PMID: 29069751 PMCID: PMC5641094 DOI: 10.18632/oncotarget.18286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/03/2017] [Indexed: 12/11/2022] Open
Abstract
Prep1 is a homeodomain transcription factor belonging to the TALE protein family. Its overexpression affects glucose metabolism in several tissues. In particular, in skeletal muscle tissue the interaction of Prep1 with its cofactor p160 impairs GLUT4 expression and glucose uptake. In this study, we show that ceramides (C2cer), a class of lipids antagonizing insulin signalling, increase the levels of Prep1 and p160 in a dose and time-dependent fashion in L6 cells and induce their association by 80%. We find that C2cer exposure inhibits insulin receptor, IRS1 and Akt phosphorylation and reduces insulin-stimulated glycogen content and glucose uptake by 1.3- and 2.1-fold, respectively. The synthetic Prep1(54-72) peptide, mimicking the Prep1 region involved in the interaction with p160, reduces in vitro Prep1-p160 binding in a dose-dependent way (IC50 = 0.20μM). In C2cer-treated L6 cells, 10μM Prep1(54-72) restores insulin signalling impaired by ceramide treatment. Prep1 overexpressing L6 cells display similar metabolic alterations observed in ceramide-treated L6 cells and the presence of Prep1(54-72) mitigates these events. All these findings suggest that disruption of the Prep1/p160 molecular interaction enhances insulin sensitivity impaired by ceramides in skeletal muscle cells and indicate this complex as an important target for type 2 diabetes.
Collapse
Affiliation(s)
- Ilaria Cimmino
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Virginia Lorenzo
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Francesca Fiory
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Nunzianna Doti
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Serena Ricci
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Antonietta Liotti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Luigi Vitagliano
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Michele Longo
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council and Interuniversity Research Centre on Bioactive Peptides, Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| |
Collapse
|
11
|
Kulebyakin K, Penkov D, Blasi F, Akopyan Z, Tkachuk V. The transcription factor Prep1 controls hepatic insulin sensitivity and gluconeogenesis by targeting nuclear localization of FOXO1. Biochem Biophys Res Commun 2016; 481:182-188. [PMID: 27815072 DOI: 10.1016/j.bbrc.2016.10.146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/29/2016] [Indexed: 10/20/2022]
Abstract
Liver plays a key role in controlling body carbohydrate homeostasis by switching between accumulation and production of glucose and this way maintaining constant level of glucose in blood. Increased blood glucose level triggers release of insulin from pancreatic β-cells. Insulin represses hepatic glucose production and increases glucose accumulation. Insulin resistance is the main cause of type 2 diabetes and hyperglycemia. Currently thiazolidinediones (TZDs) targeting transcriptional factor PPARγ are used as insulin sensitizers for treating patients with type 2 diabetes. However, TZDs are reported to be associated with cardiovascular and liver problems and stimulate obesity. Thus, it is necessary to search new approaches to improve insulin sensitivity. A promising candidate is transcriptional factor Prep1, as it was shown earlier it could affect insulin sensitivity in variety of insulin-sensitive tissues. The aim of the present study was to evaluate a possible involvement of transcriptional factor Prep1 in control of hepatic glucose accumulation and production. We created mice with liver-specific Prep1 knockout and discovered that hepatocytes derived from these mice are much more sensitive to insulin, comparing to their WT littermates. Incubation of these cells with 100 nM insulin results in almost complete inhibition of gluconeogenesis, while in WT cells this repression is only partial. However, Prep1 doesn't affect gluconeogenesis in the absence of insulin. Also, we observed that nuclear content of gluconeogenic transcription factor FOXO1 was greatly reduced in Prep1 knockout hepatocytes. These findings suggest that Prep1 may control hepatic insulin sensitivity by targeting FOXO1 nuclear stability.
Collapse
Affiliation(s)
- Konstantin Kulebyakin
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia.
| | - Dmitry Penkov
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia; IFOM - the FIRC Institute of Molecular Oncology, Via Adamello 16, Milan, 20139, Italy
| | - Francesco Blasi
- IFOM - the FIRC Institute of Molecular Oncology, Via Adamello 16, Milan, 20139, Italy
| | - Zhanna Akopyan
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia
| | - Vsevolod Tkachuk
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Department of Biochemistry and Molecular Medicine, Lomonosovsky Prospekt 31-5, Moscow, 117192, Russia
| |
Collapse
|
12
|
Penkov DN, Akopyan ZA, Kochegura TN, Egorov AD. Transcriptional control of insulin-sensitive glucose carrier Glut4 expression in adipose tissue cells. DOKL BIOCHEM BIOPHYS 2016; 467:145-9. [DOI: 10.1134/s1607672916020186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Indexed: 11/23/2022]
|
13
|
Ciccarelli M, Vastolo V, Albano L, Lecce M, Cabaro S, Liotti A, Longo M, Oriente F, Russo GL, Macchia PE, Formisano P, Beguinot F, Ungaro P. Glucose-induced expression of the homeotic transcription factor Prep1 is associated with histone post-translational modifications in skeletal muscle. Diabetologia 2016; 59:176-186. [PMID: 26453063 DOI: 10.1007/s00125-015-3774-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/10/2015] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Chronic hyperglycaemia worsens insulin resistance in individuals with type 2 diabetes. Whether this effect is contributed by epigenetic dysregulation and which genes are involved remain unclear. Prep1 (also known as Pknox1) is a gene exerting major effects on the sensitivity of the glucose transport machinery to insulin. Here, we show that dysregulation of Prep1 expression by high glucose levels is associated with histone modifications at its 5' regulatory region. METHODS We used mouse and cell models to investigate Prep1 transcriptional regulation by glucose. RESULTS Differentiated L6 skeletal muscle cells were grown in the presence of either 5.5 or 25 mmol/l glucose (normal [NG] and high glucose [HG], respectively). The HG exposure increased nuclear factor κ light chain enhancer of activated B cells (NF-κB) p65 binding and recruitment of the su(var)3-9, enhancer-of-zeste, trithorax domain-containing lysine methyltransferase 7 (SET7) histone methyltransferase and p300 acetyltransferase to the 5' region of Prep1, leading to enhanced transcription. In addition, chromatin immunoprecipitation assays revealed concomitantly increased histone H3 mono- and dimethylation and acetylation at Lys4 and Lys9/14, respectively. Skeletal muscle tissue from streptozotocin-treated diabetic mice also showed Prep1 overexpression accompanied by similarly increased recruitment of NF-κB p65 and histone modifications at the 5' region of Prep1. In these same mice, as well as in Prep1-overexpressing L6 cells, Prep1-induced recruitment of the repressor complex myocyte enhancer factor 2 (MEF2)/histone deacetylase 5 (HDAC5) at the Glut4 promoter was also increased, leading to reduced Glut4 expression. CONCLUSIONS/INTERPRETATION These studies indicate that HG exposure induces NF-κB recruitment and histone modification at the Prep1 5' region, thereby enhancing the transcription of Prep1 and repressing that of Glut4. Histone changes at the Prep1 gene may contribute to insulin resistance in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Marco Ciccarelli
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Viviana Vastolo
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Luigi Albano
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Manuela Lecce
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Serena Cabaro
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Antonietta Liotti
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Michele Longo
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Francesco Oriente
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Gian Luigi Russo
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche, Avellino, Italy
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli, 'Federico II', Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Francesco Beguinot
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Paola Ungaro
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli, 'Federico II', Naples, Italy.
- URT 'Genomica Funzionale' Istituto di Endocrinologia ed Oncologia Sperimentale, 'G. Salvatore', Consiglio Nazionale delle Ricerche, Via Sergio Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
14
|
Burgos-Ramos E, Canelles S, Rodríguez A, Gómez-Ambrosi J, Frago LM, Chowen JA, Frühbeck G, Argente J, Barrios V. Chronic central leptin infusion modulates the glycemia response to insulin administration in male rats through regulation of hepatic glucose metabolism. Mol Cell Endocrinol 2015; 415:157-72. [PMID: 26296906 DOI: 10.1016/j.mce.2015.08.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/06/2015] [Accepted: 08/09/2015] [Indexed: 10/23/2022]
Abstract
Leptin and insulin use overlapping signaling mechanisms to modify hepatic glucose metabolism, which is critical in maintaining normal glycemia. We examined the effect of an increase in central leptin and insulin on hepatic glucose metabolism and its influence on serum glucose levels. Chronic leptin infusion increased serum leptin and reduced hepatic SH-phosphotyrosine phosphatase 1, the association of suppressor of cytokine signaling 3 to the insulin receptor in liver and the rise in glycemia induced by central insulin. Leptin also decreased hepatic phosphoenolpyruvate carboxykinase levels and increased insulin's ability to phosphorylate insulin receptor substrate-1, Akt and glycogen synthase kinase on Ser9 and to stimulate glucose transporter 2 and glycogen levels. Peripheral leptin treatment reproduced some of these changes, but to a lesser extent. Our data indicate that leptin increases the hepatic response to a rise in insulin, suggesting that pharmacological manipulation of leptin targets may be of interest for controlling glycemia.
Collapse
Affiliation(s)
- Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, E-28009, Spain; IMDEA Food, CEI UAM+CSIC, Carretera de Cantoblanco 8, Madrid, E-28049, Spain
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, E-28009, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain
| | - Amaia Rodríguez
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain; Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, E-31008, Spain
| | - Javier Gómez-Ambrosi
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain; Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, E-31008, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, E-28009, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, E-28009, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain
| | - Gema Frühbeck
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain; Metabolic Research Laboratory, Clínica Universidad de Navarra, IdiSNA, Pamplona, E-31008, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, E-28009, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, E-28009, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, E-28009, Spain.
| |
Collapse
|
15
|
Raciti GA, Longo M, Parrillo L, Ciccarelli M, Mirra P, Ungaro P, Formisano P, Miele C, Béguinot F. Understanding type 2 diabetes: from genetics to epigenetics. Acta Diabetol 2015; 52:821-7. [PMID: 25841587 DOI: 10.1007/s00592-015-0741-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/14/2015] [Indexed: 12/18/2022]
Abstract
The known genetic variability (common DNA polymorphisms) does not account either for the current epidemics of type 2 diabetes or for the family transmission of this disorder. However, clinical, epidemiological and, more recently, experimental evidence indicates that environmental factors have an extraordinary impact on the natural history of type 2 diabetes. Some of these environmental hits are often shared in family groups and proved to be capable to induce epigenetic changes which alter the function of genes affecting major diabetes traits. Thus, epigenetic mechanisms may explain the environmental origin as well as the familial aggregation of type 2 diabetes much easier than common polymorphisms. In the murine model, exposure of parents to environmental hits known to cause epigenetic changes reprograms insulin sensitivity as well as beta-cell function in the progeny, indicating that certain epigenetic changes can be transgenerationally transmitted. Studies from different laboratories revealed that, in humans, lifestyle intervention modulates the epigenome and reverts environmentally induced epigenetic modifications at specific target genes. Finally, specific human epigenotypes have been identified which predict adiposity and type 2 diabetes with much greater power than any polymorphism so far identified. These epigenotypes can be recognized in easily accessible white cells from peripheral blood, indicating that, in the future, epigenetic profiling may enable effective type 2 diabetes prediction. This review discusses recent evidence from the literature supporting the immediate need for further investigation to uncover the power of epigenetics in the prediction, prevention and treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Gregory Alexander Raciti
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Michele Longo
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Luca Parrillo
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Marco Ciccarelli
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Paola Mirra
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Paola Ungaro
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Pietro Formisano
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Claudia Miele
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy
| | - Francesco Béguinot
- Dipartimento di Scienze Mediche Traslazionali, "Federico II" University of Naples Medical School, Naples, Italy.
- Istituto per l' Endocrinologia e l' Oncologia Sperimentale del C.N.R, URT "Genomica Funzionale", Via Sergio Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
16
|
Raciti GA, Nigro C, Longo M, Parrillo L, Miele C, Formisano P, Béguinot F. Personalized medicine and type 2 diabetes: lesson from epigenetics. Epigenomics 2015; 6:229-38. [PMID: 24811791 DOI: 10.2217/epi.14.10] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Similarly to genetic polymorphisms, epigenetic modifications may alter transcriptional activity and contribute to different traits of the Type 2 diabetes phenotype. The establishment of these epigenetic marks may precede diabetes onset and predict the disease. Current evidence now indicates that epigenetic differences represent markers of diabetes risk. Studies on epigenome plasticity revealed that cytokines and other metabolites, by affecting DNA methylation, may acutely reprogram gene expression and contribute to the Type 2 diabetes phenotype even in the adult life. The available evidence further indicates that epigenetic marks across the genome are subject to dynamic variations in response to environmental cues. Finally, different genes responsible for the interindividual variability in antidiabetic drug response are subjected to epigenetic regulation. Determining how specific epigenetic profiles determine diabetes is a challenging task. In the near future, the identification of epigenetic marks predictive of diabetes risk or response to treatment may offer unanticipated opportunities to personalize Type 2 diabetes management.
Collapse
Affiliation(s)
- Gregory Alexander Raciti
- Dipartimento di Scienze Mediche Traslazionali, 'Federico II' University of Naples Medical School & Istituto per l' Endocrinologia e l' Oncologia Sperimentale del CNR, Via Sergio Pansini, 5 - Naples, 80131, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Gurzov EN, Stanley WJ, Brodnicki TC, Thomas HE. Protein tyrosine phosphatases: molecular switches in metabolism and diabetes. Trends Endocrinol Metab 2015; 26:30-9. [PMID: 25432462 DOI: 10.1016/j.tem.2014.10.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are a large family of enzymes that generally oppose the actions of protein tyrosine kinases (PTKs). Genetic polymorphisms for particular PTPs are associated with altered risk of both type 1 diabetes (T1D) and type 2 diabetes (T2D). Moreover, recent evidence suggests that PTPs play crucial roles in metabolism. They can act as regulators of liver homeostasis, food intake, or immune-mediated pancreatic b cell death. In this review we describe the mechanisms by which different members of the non-receptor PTP (PTPN) family influence metabolic physiology. This 'metabolic job' of PTPs is discussed in depth and the role of these proteins in different cell types compared. Understanding the pathways regulated by PTPs will provide novel therapeutic strategies for the treatment of diabetes.
Collapse
|
18
|
Iovino S, Burkart AM, Kriauciunas K, Warren L, Hughes KJ, Molla M, Lee YK, Patti ME, Kahn CR. Genetic insulin resistance is a potent regulator of gene expression and proliferation in human iPS cells. Diabetes 2014; 63:4130-42. [PMID: 25059784 PMCID: PMC4238001 DOI: 10.2337/db14-0109] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Insulin resistance is central to diabetes and metabolic syndrome. To define the consequences of genetic insulin resistance distinct from those secondary to cellular differentiation or in vivo regulation, we generated induced pluripotent stem cells (iPSCs) from individuals with insulin receptor mutations and age-appropriate control subjects and studied insulin signaling and gene expression compared with the fibroblasts from which they were derived. iPSCs from patients with genetic insulin resistance exhibited altered insulin signaling, paralleling that seen in the original fibroblasts. Insulin-stimulated expression of immediate early genes and proliferation were also potently reduced in insulin resistant iPSCs. Global gene expression analysis revealed marked differences in both insulin-resistant iPSCs and corresponding fibroblasts compared with control iPSCs and fibroblasts. Patterns of gene expression in patients with genetic insulin resistance were particularly distinct in the two cell types, indicating dependence on not only receptor activity but also the cellular context of the mutant insulin receptor. Thus, iPSCs provide a novel approach to define effects of genetically determined insulin resistance. This study demonstrates that effects of insulin resistance on gene expression are modified by cellular context and differentiation state. Moreover, altered insulin receptor signaling and insulin resistance can modify proliferation and function of pluripotent stem cell populations.
Collapse
Affiliation(s)
- Salvatore Iovino
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Alison M Burkart
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Kristina Kriauciunas
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Laura Warren
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Katelyn J Hughes
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Michael Molla
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Youn-Kyoung Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - C Ronald Kahn
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
19
|
Risolino M, Mandia N, Iavarone F, Dardaei L, Longobardi E, Fernandez S, Talotta F, Bianchi F, Pisati F, Spaggiari L, Harter PN, Mittelbronn M, Schulte D, Incoronato M, Di Fiore PP, Blasi F, Verde P. Transcription factor PREP1 induces EMT and metastasis by controlling the TGF-β-SMAD3 pathway in non-small cell lung adenocarcinoma. Proc Natl Acad Sci U S A 2014; 111:E3775-84. [PMID: 25157139 PMCID: PMC4246949 DOI: 10.1073/pnas.1407074111] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pre-B-cell leukemia homeobox (Pbx)-regulating protein-1 (Prep1) is a ubiquitous homeoprotein involved in early development, genomic stability, insulin sensitivity, and hematopoiesis. Previously we have shown that Prep1 is a haploinsufficient tumor suppressor that inhibits neoplastic transformation by competing with myeloid ecotropic integration site 1 for binding to the common heterodimeric partner Pbx1. Epithelial-mesenchymal transition (EMT) is controlled by complex networks of proinvasive transcription factors responsive to paracrine factors such as TGF-β. Here we show that, in addition to inhibiting primary tumor growth, PREP1 is a novel EMT inducer and prometastatic transcription factor. In human non-small cell lung cancer (NSCLC) cells, PREP1 overexpression is sufficient to trigger EMT, whereas PREP1 down-regulation inhibits the induction of EMT in response to TGF-β. PREP1 modulates the cellular sensitivity to TGF-β by inducing the small mothers against decapentaplegic homolog 3 (SMAD3) nuclear translocation through mechanisms dependent, at least in part, on PREP1-mediated transactivation of a regulatory element in the SMAD3 first intron. Along with the stabilization and accumulation of PBX1, PREP1 induces the expression of multiple activator protein 1 components including the proinvasive Fos-related antigen 1 (FRA-1) oncoprotein. Both FRA-1 and PBX1 are required for the mesenchymal changes triggered by PREP1 in lung tumor cells. Finally, we show that the PREP1-induced mesenchymal transformation correlates with significantly increased lung colonization by cells overexpressing PREP1. Accordingly, we have detected PREP1 accumulation in a large number of human brain metastases of various solid tumors, including NSCLC. These findings point to a novel role of the PREP1 homeoprotein in the control of the TGF-β pathway, EMT, and metastasis in NSCLC.
Collapse
Affiliation(s)
- Maurizio Risolino
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Nadia Mandia
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy; Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy
| | - Francescopaolo Iavarone
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Leila Dardaei
- Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy
| | - Elena Longobardi
- Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy
| | - Serena Fernandez
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Francesco Talotta
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Fabrizio Bianchi
- Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy; Department of Medicine, Surgery, and Dentistry, University of Milan, 20122 Milan, Italy
| | - Federica Pisati
- Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy
| | - Lorenzo Spaggiari
- Department of Medicine, Surgery, and Dentistry, University of Milan, 20122 Milan, Italy
| | - Patrick N Harter
- Neuroscience Center, Neurological Institute (Edinger Institut), 60528 Frankfurt, Germany; and
| | - Michel Mittelbronn
- Neuroscience Center, Neurological Institute (Edinger Institut), 60528 Frankfurt, Germany; and
| | - Dorothea Schulte
- Neuroscience Center, Neurological Institute (Edinger Institut), 60528 Frankfurt, Germany; and
| | | | - Pier Paolo Di Fiore
- Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy; Department of Medicine, Surgery, and Dentistry, University of Milan, 20122 Milan, Italy
| | - Francesco Blasi
- Institute of Molecular Oncology (IFOM) of the Italian Foundation for Cancer Research (FIRC), 20139 Milan, Italy;
| | - Pasquale Verde
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy; Istituto di Ricovero e Cura a Carattere Scientifico SDN (IRCCS SDN), 80142 Naples, Italy
| |
Collapse
|
20
|
Xu E, Schwab M, Marette A. Role of protein tyrosine phosphatases in the modulation of insulin signaling and their implication in the pathogenesis of obesity-linked insulin resistance. Rev Endocr Metab Disord 2014; 15:79-97. [PMID: 24264858 DOI: 10.1007/s11154-013-9282-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Insulin resistance is a major disorder that links obesity to type 2 diabetes mellitus (T2D). It involves defects in the insulin actions owing to a reduced ability of insulin to trigger key signaling pathways in major metabolic tissues. The pathogenesis of insulin resistance involves several inhibitory molecules that interfere with the tyrosine phosphorylation of the insulin receptor and its downstream effectors. Among those, growing interest has been developed toward the protein tyrosine phosphatases (PTPs), a large family of enzymes that can inactivate crucial signaling effectors in the insulin signaling cascade by dephosphorylating their tyrosine residues. Herein we briefly review the role of several PTPs that have been shown to be implicated in the regulation of insulin action, and then focus on the Src homology 2 (SH2) domain-containing SHP1 and SHP2 enzymes, since recent reports have indicated major roles for these PTPs in the control of insulin action and glucose metabolism. Finally, the therapeutic potential of targeting PTPs for combating insulin resistance and alleviating T2D will be discussed.
Collapse
Affiliation(s)
- Elaine Xu
- Department of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Ste-Foy, Québec, Canada, G1V 4G2
| | | | | |
Collapse
|
21
|
Oriente F, Cabaro S, Liotti A, Longo M, Parrillo L, Pagano TB, Raciti GA, Penkov D, Paciello O, Miele C, Formisano P, Blasi F, Beguinot F. PREP1 deficiency downregulates hepatic lipogenesis and attenuates steatohepatitis in mice. Diabetologia 2013; 56:2713-22. [PMID: 24052111 DOI: 10.1007/s00125-013-3053-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/26/2013] [Indexed: 01/07/2023]
Abstract
AIMS/HYPOTHESIS The aim of this study was to investigate the function of Prep1 (also known as Pknox1) in hepatic lipogenesis. METHODS The hepatic lipogenesis pathway was evaluated by real-time RT-PCR and Western blot. Biochemical variables were assessed using a clinical chemistry analyser. RESULTS Serum triacylglycerols and liver expression of fatty acid synthase (FAS) were significantly decreased in Prep1 hypomorphic heterozygous (Prep1 (i/+) ) mice compared with their non-hypomorphic littermates. Upstream FAS expression, phosphorylation of protein kinase C (PKC)ζ, liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) increased in Prep1 (i/+) mice, while protein and mRNA levels of the lipid phosphatase inhibitor of PKCζ, SH2-containing inositol 5'-phosphatase 2 (SHIP2), was more than 60% reduced. Consistent with these findings, HepG2 cells transfected with Prep1 cDNA exhibited increased triacylglycerol accumulation and FAS expression, with strongly reduced PKCζ, LKB1, AMPK and ACC phosphorylation. Further experiments revealed the presence of both Prep1 and its major partner Pbx1 at the Ship2 (also known as Inppl1) promoter. PBX-regulating protein 1 (PREP1) and pre-B cell leukaemia transcription factor 1 (PBX1) enhanced Ship2 transcription. The PREP1HR mutant, which is unable to bind PBX1, exhibited no effect on Ship2 function, indicating transcriptional activation of Ship2 by the PREP1/PBX1 complex. Treatment with a methionine- and choline-deficient diet (MCDD) induced steatosis in both Prep1 (i/+) and non-hypomorphic control mice. However, alanine aminotransferase increase, intracellular triacylglycerol content and histological evidence of liver steatosis, inflammation and necrosis were significantly less evident in Prep1 (i/+) mice, indicating that Prep1 silencing protects mice from MCDD-induced steatohepatitis. CONCLUSIONS/INTERPRETATION Our results indicate that Prep1 silencing reduces lipotoxicity by increasing PKCζ/LKB1/AMPK/ACC signalling, while levels of PREP1 expression may determine the risk of steatohepatitis and its progression.
Collapse
Affiliation(s)
- Francesco Oriente
- Department of Translational Medical Sciences, 'Federico II' University of Naples and Institute of Experimental Endocrinology and Oncology, National Council of Research, Via Pansini 5, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Sharma V, Tikoo K. Stage-specific quantitative changes in renal and urinary proteome during the progression and development of streptozotocin-induced diabetic nephropathy in rats. Mol Cell Biochem 2013; 388:95-111. [PMID: 24281856 DOI: 10.1007/s11010-013-1902-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/15/2013] [Indexed: 12/31/2022]
Abstract
Diabetic nephropathy (DN) is a microvascular complication associated with diabetes causing slow deterioration of kidneys leading to end-stage renal disease. Timely intervention and diagnosis are crucial in order to ameliorate and halt the progression of DN. Current diagnosis of DN consists of urine assays for detection of microalbuminuria, which have inadequate specificity and sensitivity. Hence, there arises a need to discover stage-specific biomarkers which can aid in the early detection of DN and also in identifying the mechanisms underlying pathogenesis of DN. Therefore the present study was undertaken to identify the differentially expressed proteins in the urine and to examine the pattern of proteomic changes occurring in the rat kidneys during the course of progression of streptozotocin-induced model of DN in rats. Two-dimensional gel electrophoresis coupled to MALDI-TOF mass spectrometry was employed to identify the differentially expressed proteins under diabetic conditions. Among the identified proteins Calgranulin A and Calgranulin B appeared in the urinary proteome at the fourth week of induction of diabetes while we recorded a time-dependent decrease in the expression of major urinary protein (alpha 2u globulin) in the urine as well as kidneys of diabetic rats. Parallel monitoring of targeted proteomic changes in the renal proteome revealed an increase in histone H2B phosphorylation at serine14 along with a gradual decrease in Bcl-2 and MMP-13 expression during the course of progression and development of streptozotocin-induced DN.
Collapse
Affiliation(s)
- Vikram Sharma
- Laboratory of Chromatin Biology, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160 062, Punjab, India,
| | | |
Collapse
|
23
|
Pknox1/Prep1 regulates mitochondrial oxidative phosphorylation components in skeletal muscle. Mol Cell Biol 2013; 34:290-8. [PMID: 24216763 DOI: 10.1128/mcb.01232-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The homeodomain transcription factor Prep1 was previously shown to regulate insulin sensitivity. Our aim was to study the specific role of Prep1 for the regulation of energy metabolism in skeletal muscle. Muscle-specific ablation of Prep1 resulted in increased expression of respiratory chain subunits. This finding was consistent with an increase in mitochondrial enzyme activity without affecting mitochondrial volume fraction as assessed by electron microscopy. Metabolic phenotyping revealed no differences in daily energy expenditure or body composition. However, during treadmill exercise challenge, Prep1 ablation resulted in a higher maximal oxidative capacity and better endurance. Elevated PGC-1α expression was identified as a cause for increased mitochondrial capacity in Prep1 ablated mice. Prep1 stabilizes p160 Mybbp1a, a known inhibitor of PGC-1α activity. Thereby, p160 protein levels were significantly lower in the muscle of Prep1 ablated mice. By a chromatin immunoprecipitation-sequencing (ChIP-seq) approach, PREP1 binding sites in genes encoding mitochondrial components (e.g., Ndufs2) were identified that might be responsible for elevated proteins involved in oxidative phosphorylation (OXPHOS) in the muscle of Prep1 null mutants. These results suggest that Prep1 exhibits additional direct effects on regulation of mitochondrial proteins. We therefore conclude that Prep1 is a regulator of oxidative phosphorylation components via direct and indirect mechanisms.
Collapse
|
24
|
Penkov DN, Egorov AD, Mozgovaya MN, Tkachuk VA. Insulin resistance and adipogenesis: role of transcription and secreted factors. BIOCHEMISTRY. BIOKHIMIIA 2013; 78:8-18. [PMID: 23379555 DOI: 10.1134/s0006297913010021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Insulin stimulates carbohydrate uptake by cells and induces their conversion into lipids as a more efficient form of energy storage. Insulin resistance is associated with a decrease in glucose uptake by muscle and adipose cells and also with a decrease in glycogen synthesis on retention of glucose synthesis by liver cells. Disorders in the insulin signaling cascade on development of insulin resistance can be caused by both changes in functioning of transcriptional factors and in the secretion profile of hormone-like substances. Diacylglycerols and ceramides responsible for activation of some kinases and phosphatases can directly trigger these changes in muscle and liver cells. In adipose tissue, insulin mainly stimulates adipogenesis (adipocyte differentiation) and lipogenesis (lipid accumulation in the cells). Thus, studies on the action mechanisms of factors influencing adipogenesis can be of help for understanding the molecular mechanisms of insulin resistance.
Collapse
Affiliation(s)
- D N Penkov
- Russian Cardiology Research and Production Center, Moscow, 121552, Russia.
| | | | | | | |
Collapse
|
25
|
Xu E, Charbonneau A, Rolland Y, Bellmann K, Pao L, Siminovitch KA, Neel BG, Beauchemin N, Marette A. Hepatocyte-specific Ptpn6 deletion protects from obesity-linked hepatic insulin resistance. Diabetes 2012; 61:1949-58. [PMID: 22698917 PMCID: PMC3402325 DOI: 10.2337/db11-1502] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The protein-tyrosine phosphatase Shp1 negatively regulates insulin action on glucose homeostasis in liver and muscle, but its potential role in obesity-linked insulin resistance has not been examined. To investigate the role of Shp1 in hepatic insulin resistance, we generated hepatocyte-specific Shp1 knockout mice (Ptpn6(H-KO)), which were subjected to extensive metabolic monitoring throughout an 8-week standard chow diet (SD) or high-fat diet (HFD) feeding. We report for the first time that Shp1 expression is upregulated in metabolic tissues of HFD-fed obese mice. When compared with their Shp1-expressing Ptpn6(f/f) littermates, Ptpn6(H-KO) mice exhibited significantly lowered fasting glycemia and heightened hepatic insulin sensitivity. After HFD feeding, Ptpn6(H-KO) mice developed comparable levels of obesity as Ptpn6(f/f) mice, but they were remarkably protected from liver insulin resistance, as revealed by euglycemic clamps and hepatic insulin signaling determinations. Although Ptpn6(H-KO) mice still acquired diet-induced peripheral insulin resistance, they were less hyperinsulinemic during a glucose tolerance test because of reduced insulin secretion. Ptpn6(H-KO) mice also exhibited increased insulin clearance in line with enhanced CC1 tyrosine phosphorylation in liver. These results show that hepatocyte Shp1 plays a critical role in the development of hepatic insulin resistance and represents a novel therapeutic target for obesity-linked diabetes.
Collapse
Affiliation(s)
- Elaine Xu
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
- Department of Metabolism, Vascular and Renal Health Axis, Laval University Hospital Research Center, Québec, Québec, Canada
| | - Alexandre Charbonneau
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
- Department of Metabolism, Vascular and Renal Health Axis, Laval University Hospital Research Center, Québec, Québec, Canada
| | - Yannève Rolland
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
- Department of Metabolism, Vascular and Renal Health Axis, Laval University Hospital Research Center, Québec, Québec, Canada
| | - Kerstin Bellmann
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
- Department of Metabolism, Vascular and Renal Health Axis, Laval University Hospital Research Center, Québec, Québec, Canada
| | - Lily Pao
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Katherine A. Siminovitch
- Department of Medicine, University of Toronto, Mount Sinai Hospital Samuel Lunenfeld Research Institute, Toronto, Ontario, Canada
| | - Benjamin G. Neel
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Beauchemin
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
- Departments of Biochemistry, Medicine, and Oncology, McGill University, Montréal, Québec, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Québec, Québec, Canada
- Department of Metabolism, Vascular and Renal Health Axis, Laval University Hospital Research Center, Québec, Québec, Canada
- Corresponding author: André Marette,
| |
Collapse
|
26
|
Botta G, Passaro C, Libertini S, Abagnale A, Barbato S, Maione AS, Hallden G, Beguinot F, Formisano P, Portella G. Inhibition of autophagy enhances the effects of E1A-defective oncolytic adenovirus dl922-947 against glioma cells in vitro and in vivo. Hum Gene Ther 2012; 23:623-34. [PMID: 22475378 DOI: 10.1089/hum.2011.120] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Oncolytic viruses represent a novel therapeutic approach for aggressive tumors, such as glioblastoma multiforme, which are resistant to available treatments. Autophagy has been observed in cells infected with oncolytic viruses; however, its role in cell death/survival is unclear. To elucidate the potential therapeutic use of autophagy modulators in association with viral therapy, we analyzed autophagy induction in human glioma cell lines U373MG and U87MG infected with the oncolytic adenovirus dl922-947. dl922-947 infection triggered an autophagic cellular response, as shown by the development of acidic vesicular organelles, LC3-I→LC3-II conversion, and reduction of p62 levels. However, on infection, the Akt/mTOR/p70s6k pathway, which negatively regulates autophagy, was activated, whereas the ERK1/2 pathway, a positive regulator of autophagy, was inhibited. Accordingly, MEK inhibition by PD98059 sensitized glioma cells to dl922-947 effects, whereas autophagy induction by rapamycin protected cells from dl922-947-induced death. Treatment with two inhibitors of autophagy, chloroquine and 3-methyladenine, increased the cytotoxic effects of dl922-947 in vitro. In vivo, the growth of U87MG-induced xenografts was further reduced by adding chloroquine to the dl922-947 treatment. In conclusion, autophagy acts as a survival response in glioma cells infected with dl922-947, thus suggesting autophagy inhibitors as adjuvant/neoadjuvant drugs in oncolytic virus-based treatments.
Collapse
Affiliation(s)
- Ginevra Botta
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università Federico II, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Di Paola R, Caporarello N, Marucci A, Dimatteo C, Iadicicco C, Del Guerra S, Prudente S, Sudano D, Miele C, Parrino C, Piro S, Beguinot F, Marchetti P, Trischitta V, Frittitta L. ENPP1 affects insulin action and secretion: evidences from in vitro studies. PLoS One 2011; 6:e19462. [PMID: 21573217 PMCID: PMC3088669 DOI: 10.1371/journal.pone.0019462] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 03/30/2011] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to deeper investigate the mechanisms through which
ENPP1, a negative modulator of insulin receptor (IR) activation, plays a role on
insulin signaling, insulin secretion and eventually glucose metabolism. ENPP1
cDNA (carrying either K121 or Q121 variant) was transfected in HepG2 liver-, L6
skeletal muscle- and INS1E beta-cells. Insulin-induced IR-autophosphorylation
(HepG2, L6, INS1E), Akt-Ser473,
ERK1/2-Thr202/Tyr204 and GSK3-beta Ser9
phosphorylation (HepG2, L6), PEPCK mRNA levels (HepG2) and
2-deoxy-D-glucose uptake (L6) was studied. GLUT 4 mRNA
(L6), insulin secretion and caspase-3 activation (INS1E) were also investigated.
Insulin-induced IR-autophosphorylation was decreased in HepG2-K, L6-K, INS1E-K
(20%, 52% and 11% reduction vs. untransfected cells) and
twice as much in HepG2-Q, L6-Q, INS1E-Q (44%, 92% and 30%).
Similar data were obtained with Akt-Ser473,
ERK1/2-Thr202/Tyr204 and GSK3-beta Ser9 in
HepG2 and L6. Insulin-induced reduction of PEPCK mRNA was progressively lower in
untransfected, HepG2-K and HepG2-Q cells (65%, 54%, 23%).
Insulin-induced glucose uptake in untransfected L6 (60% increase over
basal), was totally abolished in L6-K and L6-Q cells. GLUT 4 mRNA was slightly
reduced in L6-K and twice as much in L6-Q (13% and 25% reduction
vs. untransfected cells). Glucose-induced insulin secretion was 60%
reduced in INS1E-K and almost abolished in INS1E-Q. Serum deficiency activated
caspase-3 by two, three and four folds in untransfected INS1E, INS1E-K and
INS1E-Q. Glyburide-induced insulin secretion was reduced by 50% in
isolated human islets from homozygous QQ donors as compared to those from KK and
KQ individuals. Our data clearly indicate that ENPP1, especially when the Q121
variant is operating, affects insulin signaling and glucose metabolism in
skeletal muscle- and liver-cells and both function and survival of insulin
secreting beta-cells, thus representing a strong pathogenic factor predisposing
to insulin resistance, defective insulin secretion and glucose metabolism
abnormalities.
Collapse
Affiliation(s)
- Rosa Di Paola
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
- * E-mail: (RDP); (VT); (LF)
| | - Nunzia Caporarello
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
| | - Antonella Marucci
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
| | - Claudia Dimatteo
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
| | - Claudia Iadicicco
- Dipartimento di Biologia e Patologia Cellulare
e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del CNR,
Università degli Studi di Napoli Federico II, Naples, Italy
| | - Silvia Del Guerra
- Department of Endocrinology and Metabolism,
University of Pisa, Pisa, Italy
| | - Sabrina Prudente
- IRCCS “Casa Sollievo della Sofferenza,
Mendel Laboratory”, San Giovanni Rotondo, Italy
| | - Dora Sudano
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
| | - Claudia Miele
- Dipartimento di Biologia e Patologia Cellulare
e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del CNR,
Università degli Studi di Napoli Federico II, Naples, Italy
| | - Cristina Parrino
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
| | - Salvatore Piro
- Unit of Internal Medicine, Department of
Clinical and Molecular Biomedicine, University of Catania Medical School,
Garibaldi Hospital, Catania, Italy
| | - Francesco Beguinot
- Dipartimento di Biologia e Patologia Cellulare
e Molecolare and Istituto di Endocrinologia ed Oncologia Sperimentale del CNR,
Università degli Studi di Napoli Federico II, Naples, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism,
University of Pisa, Pisa, Italy
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine
Diseases, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni
Rotondo, Italy
- IRCCS “Casa Sollievo della Sofferenza,
Mendel Laboratory”, San Giovanni Rotondo, Italy
- Department of Experimental Medicine, Sapienza
University, Rome, Italy
- * E-mail: (RDP); (VT); (LF)
| | - Lucia Frittitta
- Unit of Endocrinology, Department of Clinical
and Molecular Biomedicine, University of Catania Medical School, Garibaldi
Hospital, Catania, Italy
- * E-mail: (RDP); (VT); (LF)
| |
Collapse
|