1
|
Jin L, Yang R, Geng L, Xu A. Fibroblast Growth Factor-Based Pharmacotherapies for the Treatment of Obesity-Related Metabolic Complications. Annu Rev Pharmacol Toxicol 2023; 63:359-382. [PMID: 36100222 DOI: 10.1146/annurev-pharmtox-032322-093904] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The fibroblast growth factor (FGF) family, which comprises 22 structurally related proteins, plays diverse roles in cell proliferation, differentiation, development, and metabolism. Among them, two classical members (FGF1 and FGF4) and two endocrine members (FGF19 and FGF21) are important regulators of whole-body energy homeostasis, glucose/lipid metabolism, and insulin sensitivity. Preclinical studies have consistently demonstrated the therapeutic benefits of these FGFs for the treatment of obesity, diabetes, dyslipidemia, and nonalcoholic steatohepatitis (NASH). Several genetically engineered FGF19 and FGF21 analogs with improved pharmacodynamic and pharmacokinetic properties have been developed and progressed into various stages of clinical trials. These FGF analogs are effective in alleviating hepatic steatosis, steatohepatitis, and liver fibrosis in biopsy-confirmed NASH patients, whereas their antidiabetic and antiobesity effects are mildand vary greatly in different clinical trials. This review summarizes recent advances in biopharmaceutical development of FGF-based therapies against obesity-related metabolic complications, highlights major challenges in clinical implementation, and discusses possible strategies to overcome these hurdles.
Collapse
Affiliation(s)
- Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ranyao Yang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Leiluo Geng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China;
| |
Collapse
|
2
|
Mechanism of Jujube ( Ziziphus jujuba Mill.) Fruit in the Appetite Regulation Based on Network Pharmacology and Molecular Docking Method. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:5070086. [PMID: 35480085 PMCID: PMC9013574 DOI: 10.1155/2022/5070086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
Objective To investigate the mechanism of jujube (Ziziphus jujuba Mill.) in appetite regulation based on network pharmacology. Methods The active components and action targets of jujube were retrieved through the TCMSP and TCMID databases. GeneCards, DisGeNet, Therapeutic Target Database, and OMIM were used to screen the related targets for appetite, appetite suppression, and appetite regulation, and the intersection target of the two was selected. A protein-protein interaction (PPI) network was constructed. Important protein nodes and subnets were predicted based on the cytoHubba plug-in, and the hub gene was screened. Additionally, GO and KEGG pathway analyses were performed to obtain potential biological processes and signaling pathways of key targets. And the active ingredient-target-action pathway diagram was constructed. Results A total of 16 active components were screened from jujube, including 131 action targets related to appetite and appetite regulation. Three key targets (MAOA, MMP2, and HSPB1) were screened out by MCODE analysis. KEGG enrichment analysis was mainly enriched in neuroactive ligand-receptor interaction, serotonin-containing synapse, gap junction, cAMP signaling pathway, and dopaminergic synapse. Molecular docking results showed that the components coclaurine, (−)-catenin, (+)-stepholidine, berberine, cianidanol, coclaurine, and moupinamide in jujube had strong binding activity to the main targets (ESR1, ADRA2C, and MMP2). Conclusion Based on network pharmacology, the appetite modulating effects of jujube on multiple components, targets, and channels were explored, and the main active components of jujube were predicted to act on multiple signaling pathways to regulate appetite. The molecular docking results showed that the components in jujube had strong binding activity to the main targets, which provided new ideas and methods to further investigate the mechanisms of appetite regulation by jujube.
Collapse
|
3
|
Fan L, Ding L, Lan J, Niu J, He Y, Song L. Fibroblast Growth Factor-1 Improves Insulin Resistance via Repression of JNK-Mediated Inflammation. Front Pharmacol 2019; 10:1478. [PMID: 31866871 PMCID: PMC6906192 DOI: 10.3389/fphar.2019.01478] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/13/2019] [Indexed: 12/21/2022] Open
Abstract
Insulin resistance is associated with a greatly increased risk of type 2 diabetes. Administration of fibroblast growth factor-1 (FGF-1) resulted in a marked improvement in insulin sensitivity. However, the underlying molecular mechanism whereby FGF-1 represses insulin resistance remains largely unknown. Here, we sought to delineate the role of FGF-1 in insulin resistance with respect to its anti-inflammatory capability. In this study, we found that FGF-1 had positive effects on glucose intolerance, hepatic lipid accumulation, and insulin resistance, while it markedly repressed cytokine secretion (TNF-α and IL-6) in serum and reduced liver inflammation in diet-induced obesity (DIO) mice. Further, FGF-1 treatment significantly represses TNF-α-induced insulin resistance in vitro and in vivo. These results indicate that FGF-1 likely ameliorates insulin resistance via a mechanism that is independent of its glucose-lowering activity. Subsequent experiments demonstrated that FGF-1 ameliorated insulin resistance, and inflammation was accompanied by decreased c-Jun N-terminal kinase (JNK) signaling. In addition, it is likely that FGF-1 impedes JNK phosphorylation via blocking the transforming growth factor-β activated kinase 1 (TAK1) and TAK1 binding protein 1 (TAB1) interaction. These findings reveal that FGF-1 regulates insulin sensitivity and may represent an attractive therapeutic target for preventing the development of insulin resistance.
Collapse
Affiliation(s)
- Lei Fan
- Jinhua Hospital of Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Linchao Ding
- Jinhua Hospital of Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Junjie Lan
- School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Jianlou Niu
- School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| | - Yiling He
- Jinhua Hospital of Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Lintao Song
- School of Pharmacy, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Hultman K, Scarlett JM, Baquero AF, Cornea A, Zhang Y, Salinas CBG, Brown J, Morton GJ, Whalen EJ, Grove KL, Koegler FH, Schwartz MW, Mercer AJ. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J Comp Neurol 2019; 527:2069-2085. [PMID: 30809795 DOI: 10.1002/cne.24668] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/20/2019] [Accepted: 02/20/2019] [Indexed: 12/25/2022]
Abstract
Central activation of fibroblast growth factor (FGF) receptors regulates peripheral glucose homeostasis and reduces food intake in preclinical models of obesity and diabetes. The current work was undertaken to advance our understanding of the receptor expression, as sites of ligand action by FGF19, FGF21, and FGF1 in the mammalian brain remains unresolved. Recent advances in automated RNAscope in situ hybridization and droplet digital PCR (ddPCR) technology allowed us to interrogate central FGFR/beta klotho (Klb) system at the cellular level in the mouse, with relevant comparisons to nonhuman primate and human brain. FGFR1-3 gene expression was broadly distributed throughout the CNS in Mus musculus, with FGFR1 exhibiting the greatest heterogeneity. FGFR4 expression localized only in the medial habenula and subcommissural organ of mice. Likewise, Klb mRNA was restricted to the suprachiasmatic nucleus (SCh) and select midbrain and hindbrain nuclei. ddPCR in the rodent hypothalamus confirmed that, although expression levels are indeed low for Klb, there is nonetheless a bonafide subpopulation of Klb+ cells in the hypothalamus. In NHP and human midbrain and hindbrain, Klb + cells are quite rare, as is expression of FGFR4. Collectively, these data provide the most robust central map of the FGFR/Klb system to date and highlight central regions that may be of critical importance to assess central ligand effects with pharmacological dosing, such as the putative interactions between the endocrine FGFs and FGFR1/Klb, or FGF19 with FGFR4.
Collapse
Affiliation(s)
| | - Jarrad M Scarlett
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington.,Department of Pediatric Gastroenterology & Hepatology, Seattle Children's Hospital, Seattle, Washington
| | - Arian F Baquero
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Anda Cornea
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Yu Zhang
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | | | - Jenny Brown
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Gregory J Morton
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Erin J Whalen
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Kevin L Grove
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Frank H Koegler
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| | - Michael W Schwartz
- Diabetes & Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington
| | - Aaron J Mercer
- Novo Nordisk Research Center Seattle, Inc., Seattle, Washington
| |
Collapse
|
5
|
Abstract
A hypercaloric diet combined with a sedentary lifestyle is a major risk factor for the development of insulin resistance, type 2 diabetes mellitus (T2DM) and associated comorbidities. Standard treatment for T2DM begins with lifestyle modification, and includes oral medications and insulin therapy to compensate for progressive β-cell failure. However, current pharmaceutical options for T2DM are limited in that they do not maintain stable, durable glucose control without the need for treatment intensification. Furthermore, each medication is associated with adverse effects, which range from hypoglycaemia to weight gain or bone loss. Unexpectedly, fibroblast growth factor 1 (FGF1) and its low mitogenic variants have emerged as potentially safe candidates for restoring euglycaemia, without causing overt adverse effects. In particular, a single peripheral injection of FGF1 can lower glucose to normal levels within hours, without the risk of hypoglycaemia. Similarly, a single intracerebroventricular injection of FGF1 can induce long-lasting remission of the diabetic phenotype. This Review discusses potential mechanisms by which centrally administered FGF1 improves central glucose-sensing and peripheral glucose uptake in a sustained manner. Specifically, we explore the potential crosstalk between FGF1 and glucose-sensing neuronal circuits, hypothalamic neural stem cells and synaptic plasticity. Finally, we highlight therapeutic considerations of FGF1 and compare its metabolic actions with FGF15 (rodents), FGF19 (humans) and FGF21.
Collapse
Affiliation(s)
- Emanuel Gasser
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Christopher P Moutos
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
- College of Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, Arkansas 72205, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
6
|
Scarlett JM, Rojas JM, Matsen ME, Kaiyala KJ, Stefanovski D, Bergman RN, Nguyen HT, Dorfman MD, Lantier L, Wasserman DH, Mirzadeh Z, Unterman TG, Morton GJ, Schwartz MW. Central injection of fibroblast growth factor 1 induces sustained remission of diabetic hyperglycemia in rodents. Nat Med 2016; 22:800-6. [PMID: 27213816 PMCID: PMC4938755 DOI: 10.1038/nm.4101] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 04/08/2016] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes (T2D) is among the most common and costly disorders worldwide. The goal of current medical management for T2D is to transiently ameliorate hyperglycemia through daily dosing of one or more antidiabetic drugs. Hypoglycemia and weight gain are common side effects of therapy, and sustained disease remission is not obtainable with nonsurgical approaches. On the basis of the potent glucose-lowering response elicited by activation of brain fibroblast growth factor (FGF) receptors, we explored the antidiabetic efficacy of centrally administered FGF1, which, unlike other FGF peptides, activates all FGF receptor subtypes. We report that a single intracerebroventricular injection of FGF1 at a dose one-tenth of that needed for antidiabetic efficacy following peripheral injection induces sustained diabetes remission in both mouse and rat models of T2D. This antidiabetic effect is not secondary to weight loss, does not increase the risk of hypoglycemia, and involves a novel and incompletely understood mechanism for increasing glucose clearance from the bloodstream. We conclude that the brain has an inherent potential to induce diabetes remission and that brain FGF receptors are potential pharmacological targets for achieving this goal.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/metabolism
- Animals
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Blotting, Western
- Body Composition
- Brain/drug effects
- Brain/metabolism
- Carbon Radioisotopes
- Deoxyglucose
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat
- Disease Models, Animal
- Ependymoglial Cells/drug effects
- Ependymoglial Cells/metabolism
- Fibroblast Growth Factor 1/pharmacology
- Forkhead Box Protein O1/genetics
- Glucose Tolerance Test
- Heart/drug effects
- Heat-Shock Proteins/drug effects
- Heat-Shock Proteins/metabolism
- Hyperglycemia/metabolism
- Hypothalamus/cytology
- Hypothalamus/drug effects
- Hypothalamus/metabolism
- Injections, Intraventricular
- Liver/metabolism
- Male
- Mice
- Mice, Knockout
- Mice, Obese
- Molecular Chaperones
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Myocardium/metabolism
- Neoplasm Proteins/drug effects
- Neoplasm Proteins/metabolism
- Proto-Oncogene Proteins c-fos/drug effects
- Proto-Oncogene Proteins c-fos/metabolism
- Rats
- Rats, Zucker
- Real-Time Polymerase Chain Reaction
- Receptor, Insulin/antagonists & inhibitors
- Receptor, Insulin/genetics
- Remission Induction
Collapse
Affiliation(s)
- Jarrad M Scarlett
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, Washington, USA
| | - Jennifer M Rojas
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Miles E Matsen
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Karl J Kaiyala
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, Washington, USA
| | - Darko Stefanovski
- New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hong T Nguyen
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Mauricio D Dorfman
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt School of Medicine, Nashville, Tennessee, USA
| | - Zaman Mirzadeh
- Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Terry G Unterman
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
- Medical Service, Jesse Brown Virginia Medical Center, Chicago, Illinois, USA
| | - Gregory J Morton
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Michael W Schwartz
- Diabetes and Obesity Center of Excellence, Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Nies VJM, Sancar G, Liu W, van Zutphen T, Struik D, Yu RT, Atkins AR, Evans RM, Jonker JW, Downes MR. Fibroblast Growth Factor Signaling in Metabolic Regulation. Front Endocrinol (Lausanne) 2015; 6:193. [PMID: 26834701 PMCID: PMC4718082 DOI: 10.3389/fendo.2015.00193] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/25/2015] [Indexed: 12/22/2022] Open
Abstract
The prevalence of obesity is a growing health problem. Obesity is strongly associated with several comorbidities, such as non-alcoholic fatty liver disease, certain cancers, insulin resistance, and type 2 diabetes, which all reduce life expectancy and life quality. Several drugs have been put forward in order to treat these diseases, but many of them have detrimental side effects. The unexpected role of the family of fibroblast growth factors in the regulation of energy metabolism provides new approaches to the treatment of metabolic diseases and offers a valuable tool to gain more insight into metabolic regulation. The known beneficial effects of FGF19 and FGF21 on metabolism, together with recently discovered similar effects of FGF1 suggest that FGFs and their derivatives carry great potential as novel therapeutics to treat metabolic conditions. To facilitate the development of new therapies with improved targeting and minimal side effects, a better understanding of the molecular mechanism of action of FGFs is needed. In this review, we will discuss what is currently known about the physiological roles of FGF signaling in tissues important for metabolic homeostasis. In addition, we will discuss current concepts regarding their pharmacological properties and effector tissues in the context of metabolic disease. Also, the recent progress in the development of FGF variants will be reviewed. Our goal is to provide a comprehensive overview of the current concepts and consensuses regarding FGF signaling in metabolic health and disease and to provide starting points for the development of FGF-based therapies against metabolic conditions.
Collapse
Affiliation(s)
- Vera J. M. Nies
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gencer Sancar
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Weilin Liu
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Tim van Zutphen
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Dicky Struik
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ruth T. Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Annette R. Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ronald M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Johan W. Jonker
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Johan W. Jonker, ; Michael Robert Downes,
| | - Michael Robert Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- *Correspondence: Johan W. Jonker, ; Michael Robert Downes,
| |
Collapse
|
8
|
Suh JM, Jonker JW, Ahmadian M, Goetz R, Lackey D, Osborn O, Huang Z, Liu W, Yoshihara E, van Dijk T, Havinga R, Fan W, Yin YQ, Yu RT, Liddle C, Atkins AR, Olefsky JM, Mohammadi M, Downes M, Evans RM. Endocrinization of FGF1 produces a neomorphic and potent insulin sensitizer. Nature 2014; 513:436-9. [PMID: 25043058 PMCID: PMC4184286 DOI: 10.1038/nature13540] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 05/29/2014] [Indexed: 01/18/2023]
Abstract
Fibroblast growth factor 1 (FGF1) is an autocrine/paracrine regulator whose binding to heparan sulphate proteoglycans effectively precludes its circulation. Although FGF1 is known as a mitogenic factor, FGF1 knockout mice develop insulin resistance when stressed by a high-fat diet, suggesting a potential role in nutrient homeostasis. Here we show that parenteral delivery of a single dose of recombinant FGF1 (rFGF1) results in potent, insulin-dependent lowering of glucose levels in diabetic mice that is dose-dependent but does not lead to hypoglycaemia. Chronic pharmacological treatment with rFGF1 increases insulin-dependent glucose uptake in skeletal muscle and suppresses the hepatic production of glucose to achieve whole-body insulin sensitization. The sustained glucose lowering and insulin sensitization attributed to rFGF1 are not accompanied by the side effects of weight gain, liver steatosis and bone loss associated with current insulin-sensitizing therapies. We also show that the glucose-lowering activity of FGF1 can be dissociated from its mitogenic activity and is mediated predominantly via FGF receptor 1 signalling. Thus we have uncovered an unexpected, neomorphic insulin-sensitizing action for exogenous non-mitogenic human FGF1 with therapeutic potential for the treatment of insulin resistance and type 2 diabetes.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Body Weight/drug effects
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat
- Dose-Response Relationship, Drug
- Fibroblast Growth Factor 1/administration & dosage
- Fibroblast Growth Factor 1/adverse effects
- Fibroblast Growth Factor 1/pharmacology
- Glucose/metabolism
- Glucose Tolerance Test
- Humans
- Insulin/metabolism
- Insulin Resistance
- Liver/drug effects
- Liver/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Obese
- Mitogens/pharmacology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
Collapse
Affiliation(s)
- Jae Myoung Suh
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Johan W. Jonker
- Center for Liver, Digestive and Metabolic Diseases, Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Maryam Ahmadian
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Regina Goetz
- Department of Pharmacology, New York University School of Medicine, New York, New York 10016, USA
| | - Denise Lackey
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| | - Olivia Osborn
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| | - Zifeng Huang
- Department of Pharmacology, New York University School of Medicine, New York, New York 10016, USA
| | - Weilin Liu
- Center for Liver, Digestive and Metabolic Diseases, Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Eiji Yoshihara
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Theo van Dijk
- Center for Liver, Digestive and Metabolic Diseases, Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Rick Havinga
- Center for Liver, Digestive and Metabolic Diseases, Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Weiwei Fan
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Yun-Qiang Yin
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Ruth T. Yu
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Christopher Liddle
- The Storr Liver Unit, Westmead Millennium Institute and University of Sydney, Westmead Hospital, Westmead, New South Wales 2145, Australia
| | - Annette R. Atkins
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Jerrold M. Olefsky
- Department of Medicine, Division of Endocrinology and Metabolism, University of California at San Diego, La Jolla, California 92093, USA
| | - Moosa Mohammadi
- Department of Pharmacology, New York University School of Medicine, New York, New York 10016, USA
| | - Michael Downes
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Ronald M. Evans
- GeneExpression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
9
|
Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. A simple method to obtain pure cultures of multiciliated ependymal cells from adult rodents. Histochem Cell Biol 2012; 139:205-20. [PMID: 22878526 DOI: 10.1007/s00418-012-1008-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2012] [Indexed: 11/25/2022]
Abstract
Ependymal cells form an epithelium lining the ventricular cavities of the vertebrate brain. Numerous methods to obtain primary culture ependymal cells have been developed. Most of them use foetal or neonatal rat brain and the few that utilize adult brain hardly achieve purity. Here, we describe a simple and novel method to obtain a pure non-adherent ependymal cell culture from explants of the striatal and septal walls of the lateral ventricles. The combination of a low incubation temperature followed by a gentle enzymatic digestion allows the detachment of most of the ependymal cells from the ventricular wall in a period of 6 h. Along with ependymal cells, a low percentage (less than 6 %) of non-ependymal cells also detaches. However, they do not survive under two restrictive culture conditions: (1) a simple medium (alpha-MEM with glucose) without any supplement; and (2) a low density of 1 cell/µl. This purification method strategy does not require cell labelling with antibodies and cell sorting, which makes it a simpler and cheaper procedure than other methods previously described. After a period of 48 h, only ependymal cells survive such conditions, revealing the remarkable survival capacity of ependymal cells. Ependymal cells can be maintained in culture for up to 7-10 days, with the best survival rates obtained in Neurobasal supplemented with B27 among the tested media. After 7 days in culture, ependymal cells lose most of the cilia and therefore the mobility, while acquiring radial glial cell markers (GFAP, BLBP, GLAST). This interesting fact might indicate a reprogramming of the cell identity.
Collapse
Affiliation(s)
- J M Grondona
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Campus de Teatinos, Universidad de Málaga, 29071 Málaga, Spain.
| | | | | | | |
Collapse
|
10
|
Hebb MO, Myers TL, Clarke DB. Enhanced expression of heat shock protein 27 is correlated with axonal regeneration in mature retinal ganglion cells. Brain Res 2006; 1073-1074:146-50. [PMID: 16476415 DOI: 10.1016/j.brainres.2005.12.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 12/01/2005] [Accepted: 12/05/2005] [Indexed: 11/27/2022]
Abstract
The small heat shock protein, Hsp27, promotes axonal regeneration in peripheral neurons; however, an analogous role in the central nervous system has not been described. This study examined the relationship between Hsp27 expression and regeneration in mature retinal ganglion cells (RGCs). Adult rat optic nerves were transected and exposed to peripheral nerve autografts to stimulate regeneration of cut RGC axons. There was a five-fold increase in the Hsp27-positive fraction of RGCs that extended new axons into the graft when compared with those that survived injury but did not regenerate (30% versus 6% respectively, P = 0.001). Hsp27 protein was located throughout somata and neuritic processes, and there was a significant positive correlation between Hsp27 expression and axonal regeneration in injured neurons (R = 0.92, P < 0.0001). These findings are consistent with the growth-associated role of Hsp27 demonstrated in peripheral neurons and suggest that Hsp27 may mediate similar physiological functions in the central nervous system.
Collapse
Affiliation(s)
- Matthew O Hebb
- Department of Anatomy & Neurobiology, Dalhousie University, Halifax, NS, Canada B3H 4H7
| | | | | |
Collapse
|
11
|
Gautron L, Mingam R, Moranis A, Combe C, Layé S. Influence of feeding status on neuronal activity in the hypothalamus during lipopolysaccharide-induced anorexia in rats. Neuroscience 2005; 134:933-46. [PMID: 16039792 DOI: 10.1016/j.neuroscience.2005.03.063] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2004] [Revised: 03/20/2005] [Accepted: 03/30/2005] [Indexed: 10/25/2022]
Abstract
Fasting attenuates disease-associated anorexia, but the mechanisms underlying this effect are not well understood. In the present study, we investigated the extent to which a 48 h fast alters hypothalamic neuronal activity in response to the anorectic effects of lipopolysaccharide in rats. Male rats were fed ad libitum or fasted, and were injected with i.p. saline or lipopolysaccharide (250 microg/kg). Immunohistochemistry for Fos protein was used to visualize neuronal activity in response to lipopolysaccharide within selected hypothalamic feeding regulatory nuclei. Additionally, food intake, body weight, plasma interleukin-1 and leptin levels, and the expression of mRNA for appetite-related neuropeptides (neuropeptide Y, proopiomelanocortin and cocaine-amphetamine-regulated transcript) were measured in a time-related manner. Our data show that the pattern of lipopolysaccharide-induced Fos expression was similar in most hypothalamic nuclei whatever the feeding status. However, we observed that fasting significantly reduced lipopolysaccharide-induced Fos expression in the paraventricular nucleus, in association with an attenuated lipopolysaccharide-induced anorexia and body weight loss. Moreover, lipopolysaccharide reduced fasting-induced Fos expression in the perifornical area of the lateral hypothalamus. Lipopolysaccharide-induced circulating levels of interleukin-1 were similar across feeding status. Finally, fasting, but not lipopolysaccharide, affected circulating level of leptin and appetite-related neuropeptides expression in the arcuate nucleus. Together, our data show that fasting modulates lipopolysaccharide-induced anorexia and body weight loss in association with neural changes in specific hypothalamic nuclei.
Collapse
Affiliation(s)
- L Gautron
- Laboratoire des Régulations Neuroendocriniennes, EA 2972, Université Bordeaux I, 33400 Talence, France
| | | | | | | | | |
Collapse
|
12
|
Iwadate Y, Inoue M, Saegusa T, Tokusumi Y, Kinoh H, Hasegawa M, Tagawa M, Yamaura A, Shimada H. Recombinant Sendai virus vector induces complete remission of established brain tumors through efficient interleukin-2 gene transfer in vaccinated rats. Clin Cancer Res 2005; 11:3821-7. [PMID: 15897582 DOI: 10.1158/1078-0432.ccr-04-1485] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Sendai virus (SeV), a murine parainfluenza virus type I, replicates independent of cellular genome and directs high-level gene expressions when used as a viral vector. We constructed a nontransmissible recombinant SeV vector by deleting the matrix (M) and fusion (F) genes from its genome (SeV/DeltaMDeltaF) to enhance its safety. We also estimated the therapeutic efficacy of the novel vector system against a rat glioblastoma model. EXPERIMENTAL DESIGN We administered the recombinant SeV vector carrying the lacZ gene or the human interleukin-2 (hIL-2) gene into established 9L brain tumors in vivo simultaneous with peripheral vaccination using irradiated 9L cells. Sequential monitoring with magnetic resonance imaging was used to evaluate the therapeutic efficacy. RESULTS We found extensive transduction of the lacZ gene into the brain tumors and confirmed sufficient amounts of interleukin 2 (IL-2) production by hIL2-SeV/DeltaMDeltaF both in vitro and in vivo. The magnetic resonance imaging study showed that the intracerebral injection of hIL2-SeV/DeltaMDeltaF brought about significant reduction of the tumor growth, including complete elimination of the established brain tumors. The (51)Cr release assay showed that significant amounts of 9L-specific cytotoxic T cells were induced by the peripheral vaccination. Immunohistochemical analysis revealed that CD4(+) T cells and CD8(+) T cells were abundantly infiltrated in the target tumors. CONCLUSION The present results show that the recombinant nontransmissible SeV vector provides efficient in vivo gene transfer that induces significant regression of the established brain tumors and suggest that it will be a safe and useful viral vector for the clinical practice of glioma gene therapy.
Collapse
Affiliation(s)
- Yasuo Iwadate
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Goldbaum O, Richter-Landsberg C. Proteolytic stress causes heat shock protein induction, tau ubiquitination, and the recruitment of ubiquitin to tau-positive aggregates in oligodendrocytes in culture. J Neurosci 2004; 24:5748-57. [PMID: 15215297 PMCID: PMC6729227 DOI: 10.1523/jneurosci.1307-04.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2003] [Revised: 05/10/2004] [Accepted: 05/10/2004] [Indexed: 11/21/2022] Open
Abstract
Molecular chaperones and the ubiquitin-proteasome system are participants in the defense against unfolded proteins and provide an effective protein quality control system that is essential for cellular functions and survival. Ubiquitinated tau-positive inclusion bodies containing the small heat shock protein alphaB-crystallin in oligodendrocytes are consistent features of a variety of neurodegenerative diseases, and defects in the proteasome system might contribute to the aggregation process. Oligodendrocytes, the myelin-forming cells of the CNS, are specifically sensitive to stress situations. Here we can show that in cultured rat brain oligodendrocytes proteasomal inhibition by MG-132 or lactacystin caused apoptotic cell death and the induction of heat shock proteins in a time- and concentration-dependent manner. Specifically, alphaB-crystallin was upregulated, and ubiquitinated proteins accumulated. After incubation with MG-132 the tau was dephosphorylated, which enhanced its microtubule-binding capacity. Proteasomal inhibition led to ubiquitination of tau and its association with alphaB-crystallin and to the occurrence of thioflavine S-positive aggregates in the oligodendroglial cytoplasm. These aggregates were positive for tau and also contained ubiquitin and alphaB-crystallin; hence they resembled the glial cytoplasmic inclusions observed in white matter disease and frontotemporal dementias with parkinsonism linked to chromosome 17 (FTDP-17). In summary, the data underscore the specific sensitivity of oligodendrocytes to stress situations and point to a causal relationship of proteasomal impairment and inclusion body formation.
Collapse
Affiliation(s)
- Olaf Goldbaum
- Department of Biology, Molecular Neurobiology, University of Oldenburg, D-26111 Oldenburg, Germany
| | | |
Collapse
|
14
|
Prima V, Tennant M, Gorbatyuk OS, Muzyczka N, Scarpace PJ, Zolotukhin S. Differential modulation of energy balance by leptin, ciliary neurotrophic factor, and leukemia inhibitory factor gene delivery: microarray deoxyribonucleic acid-chip analysis of gene expression. Endocrinology 2004; 145:2035-45. [PMID: 14715713 DOI: 10.1210/en.2003-1376] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Most obese animal models, whether associated with genetic, diet-induced, or age-related obesity, display pronounced leptin resistance, rendering leptin supplement therapy ineffective in treating obesity. Ciliary neurotrophic factor (CNTF) has been recently used to invoke leptin-like signaling pathways, thereby circumventing leptin resistance. In the current study, we characterize immediate and long-term molecular events in the hypothalamus of rats exposed to the sustained ectopic expression of leptin, CNTF, or leukemia inhibitory factor, another neurocytokine of IL-6 family, all delivered centrally via a viral vector. The respective transgene-encoded ligands induced similar but not identical metabolic responses as assessed by the reduction in body weight gain and changes in food intake. To define molecular mechanisms of weight-reducing and anorexigenic action of cytokines, we have analyzed the gene expression profiles of 1300 brain-specific genes in the hypothalami of normal rats subjected to the prolonged cytokine action for 10 wk. We present evidence that constitutive expression of cytokines in the brain induces changes in gene expression characteristic of chronic inflammation leading to either temporal weight reduction (CNTF) or severe cachexia (leukemia inhibitory factor). Our results convey a cautionary note regarding potential use of the tested cytokines in therapeutic applications.
Collapse
Affiliation(s)
- V Prima
- Department of Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida, Gainesville, Florida 32610-0266, USA
| | | | | | | | | | | |
Collapse
|
15
|
Costentin J. Éléments de physiologie et de neurobiologie de la prise alimentaire. ANNALES PHARMACEUTIQUES FRANÇAISES 2004; 62:92-102. [PMID: 15107726 DOI: 10.1016/s0003-4509(04)94287-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Many methods and techniques have accumulated a considerable mass of data about the mechanisms which control food intake and energetic loss. After a presentation of the main experimental approaches in this respect, the most relevant signals sent by peripheral organs to the central nervous system are presented: Glucose, triglycerides; from adipocytes: Leptin; from pancreas: Insulin, pancreatic polypeptide, amylin, enterostatin; from digestive tract: Ghrelin, cholecystokinin, peptide Y Y 3-36. Then are considered, especially at the hypothalamic level, their interpretation by neurones whose transmitters are either neuropeptides such as: Neuropeptide Y, Agouti Related Peptide, Cocaine/Amphetamine Regulated Transcript, Melanin Concentrating Hormone, alpha Melanocyte Stimulating Hormone, orexins/hypocretins, octadecaneuropeptide, nociceptin/orphanin FQ, opioid peptides, Interleukin 1, galanin, urocortin 2, Neurotrophic ciliary factor, or monoamines such as: Glutamate, dopamine, Norepinephrine, serotonine, GABA, histamine, acetylcholine. In a last part are considered the likely relationships existing between feeding, pleasure and addiction, centered on dopamine transmission in the nucleus accumbens. After this brief synopsis one should not be surprised that this so complex system which regulates feeding may be affected by various disorders; however one may be amazed by such a scarcity of drugs to influence it; in any case, many new pharmacological strategies can be expected in the future.
Collapse
Affiliation(s)
- J Costentin
- Unité de Neuropsychopharmacologie, U.M.R. 6036 CNRS, Faculté de Médecine & Pharmacie, 22, Bd Gambetta, F76183 Rouen Cedex 1
| |
Collapse
|
16
|
Bitzer M, Armeanu S, Lauer UM, Neubert WJ. Sendai virus vectors as an emerging negative-strand RNA viral vector system. J Gene Med 2003; 5:543-53. [PMID: 12825193 DOI: 10.1002/jgm.426] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The power to manipulate the genome of negative-strand RNA viruses, including the insertion of additional non-viral genes, has led to the development of a new class of viral vectors for gene transfer approaches. The murine parainfluenza virus type I, or Sendai virus (SeV), has emerged as a prototype virus of this vector group, being employed in numerous in vitro as well as animal studies over the last few years. Extraordinary features of SeV are the remarkably brief contact time that is necessary for cellular uptake, a strong but adjustable expression of foreign genes, efficient infection in the respiratory tract despite a mucus layer, transduction of target cells being independent of the cell cycle, and an exclusively cytoplasmic replication cycle without any risk of chromosomal integration. In this review we describe the current knowledge of Sendai virus vector (SeVV) development as well as the results of first-generation vector applications under both in vitro and in vivo conditions. So far, Sendai virus vectors have been identified to be a highly efficient transduction tool for a broad range of different tissues and applications. Future directions in vector design and development are discussed.
Collapse
Affiliation(s)
- Michael Bitzer
- Internal Medicine I, Medical University Clinic Tübingen, 72076 Tübingen, Germany.
| | | | | | | |
Collapse
|
17
|
Hirata K, He J, Hirakawa Y, Liu W, Wang S, Kawabuchi M. HSP27 is markedly induced in Schwann cell columns and associated regenerating axons. Glia 2003; 42:1-11. [PMID: 12594732 DOI: 10.1002/glia.10105] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It is well known that regenerating axons enter Schwann cell (SC) columns, within which they grow to reinnervate the appropriate targets. The current study detected a marked induction of a 27-kDa heat shock protein (HSP27) in the SC columns of crush-injured rat sciatic nerves. Immunohistochemical studies showed the first appearance of strong HSP27-immunoreactive linear structures in the proximal stump near an injury site 7 h after an operation. The HSP27-immunoreactive linear structures crossed the injury site to the distal stump 2 days after the operation. They then extended in a more proximal and more distal direction and were found to have propagated through the entire length of the nerve 1 week after the operation. This pattern of expression was maintained until 3 weeks after the operation. Double-immunofluorescent labeling and confocal laser microscopy confirmed that the linear structures consisted of SC columns and associated multiple axons. The HSP27-immunoreactive SC columns expressed glial fibrillary acidic protein, but not S-100 protein. Electron microscopy and immunoelectron microscopy demonstrated that reactive Schwann cells (SCs) and the associated axons with an outgrowing profile exhibited a strong immunoreactivity to HSP27, with the former containing a greater number of bundles of intermediate filaments. It is suggested that HSP27 may play an essential role in axonal outgrowth, especially by contributing to cytoskeletal dynamics in SCs.
Collapse
Affiliation(s)
- Kazuho Hirata
- Department of Anatomy and Cell Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | |
Collapse
|
18
|
Armeanu S, Ungerechts G, Bernloehr C, Bossow S, Gregor M, Neubert WJ, Lauer UM, Bitzer M. Cell cycle independent infection and gene transfer by recombinant Sendai viruses. J Virol Methods 2003; 108:229-33. [PMID: 12609691 DOI: 10.1016/s0166-0934(02)00280-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A common problem for viral vectors in the field of somatic gene therapy is the dependence of an efficient cellular transduction on the cell cycle phase of target cells. An optimized viral vector system should therefore transduce cells in different cell cycle phases equally to improve transduction efficiencies. Recent observations that recombinant Sendai viruses (SeV) can infect a broad range of different tissues suggested SeV to be a good candidate for future gene therapeutic strategies in which dividing and non-dividing cells have to be reached. However, detailed data on the influence of distinct cell cycle phases on the infection of SeV or related viruses are missing. We report that synchronization of NIH 3T3 cells as well as contact inhibition of human fibroblast cells did not exhibit any negative influence on SeV infection rates. Furthermore, different attractive target tissues like human umbilical cord derived cells or primary human hepatocytes can be reached by SeV efficiently. As an important information for further cell cycle studies of paramyxoviruses we discovered surprisingly that the DNA polymerase inhibitor aphidicolin (induces a G(1)/M arrest) functions as an inhibitor of SeV but not of an adenoviral expression vector. In conclusion, the results demonstrate SeV based vector particles to be an ideal tool to reach equally cells coexisting in different cell cycle phases.
Collapse
Affiliation(s)
- Sorin Armeanu
- Internal Medicine I, University Clinic Tübingen, D-72076, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Bitzer M, Ungerechts G, Bossow S, Graepler F, Sedlmeier R, Armeanu S, Bernloehr C, Spiegel M, Gross CD, Gregor M, Neubert WJ, Lauer UM. Negative-strand RNA viral vectors: intravenous application of Sendai virus vectors for the systemic delivery of therapeutic genes. Mol Ther 2003; 7:210-7. [PMID: 12597909 DOI: 10.1016/s1525-0016(02)00052-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Treatment by gene replacement is critical in the field of gene therapy. Suitable vectors for the delivery of therapeutic genes have to be generated and tested in preclinical settings. Recently, extraordinary features for a local gene delivery by Sendai virus vectors (SeVV) have been reported for different tissues. Here we show that direct intravenous application of SeVV in mice is not only feasible and safe, but it results in the secretion of therapeutic proteins to the circulation, for example, human clotting Factor IX (hFIX). In vitro characterization of first-generation SeVV demonstrated that secreted amounts of hFIX were at least comparable to published results for retroviral or adeno-associated viral vectors. Furthermore, as a consideration for application in humans, SeVV transduction led to efficient hFIX synthesis in primary human hepatocytes, and SeVV-encoded hFIX proteins could be shown to be functionally active in the human clotting cascade. In conclusion, our investigations demonstrate for the first time that intravenous administration of negative-strand RNA viral vectors may become a useful tool for the wide area of gene replacement requirements.
Collapse
Affiliation(s)
- Michael Bitzer
- Internal Medicine I, University Clinic Tübingen, D-72076 Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
We have previously reported that a specialized subpopulation of astrocytes in the arcuate nucleus of the hypothalamus show an unusually intense immunoreactivity for brain fatty acid binding protein (bFABP). Since bFABP has been shown to regulate the activity of an enzyme, fatty acid synthase, that has a potent influence upon the regulation of feeding by the hypothalamus, it was of interest to determine if bFABP + astrocytes are positioned to potentially influence the activity of feeding-regulating neurones. In this study, we examined the anatomical relationship between specialized arcuate astrocytes immunoreactive for bFABP and feeding-regulating neurones that are responsive to leptin and which are immunoreactive for the transcription factor STAT3. The results show that both cell types are abundant in the arcuate nucleus of the hypothalamus and are frequently closely adjacent to each other. This study provides an anatomical basis for the possibility that specialized arcuate astrocytes regulate the function of leptin-sensitive, feeding-regulating neurones of the arcuate nucleus.
Collapse
Affiliation(s)
- John K Young
- Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA.
| |
Collapse
|
21
|
Suzuki S, Li AJ, Ikemoto M, Imamura T. Expression of tenascin-C long isoforms is induced in the hypothalamus by FGF-1. Neuroreport 2002; 13:1041-5. [PMID: 12060805 DOI: 10.1097/00001756-200206120-00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Fibroblast growth factor (FGF)-1 modulates various brain functions, such as the hypothalamic control of feeding. In the rat, we examined the effect of intracerebroventricularly infused FGF-1 on the hypothalamic expression of tenascin-C, a selective mediator of neuron-glial interaction. In situ hybridization revealed little tenascin-C mRNA expression in control brains, but greatly increased expression in ependymal cells around the third ventricle in the FGF-1-infused rats. Reverse transcription-linked PCR analysis of hypothalamic mRNA revealed an FGF-1-induced expression not of the shortest isoform of tenascin-C, but of the long isoforms (with additional fibronectin type III-domain insertions). Quantitative analysis by real time PCR revealed that this induction was transient and dose-dependent. Specific modulation of hypothalamic neuron-glial interactions by tenascin-C may mediate FGF-1-induced feeding suppression.
Collapse
Affiliation(s)
- Seigo Suzuki
- Gene Discovery Research Center, National Institute of Advanced Industrial Science and Technology (AIST), AIST Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | | | | | | |
Collapse
|
22
|
Abstract
The high incidence of obesity, its multifactorial nature, the complexity and lack of knowledge of the bodyweight control system, and the scarcity of adequate therapeutics have fuelled anti-obesity drug development during a considerable number of years. Irrespective of the efforts invested by researchers and companies, few products have reached a minimum level of effectiveness, and even fewer are available in medical practice. As a consequence of anti-obesity research, our knowledge of the bodyweight control system increased but, despite this, the pharmacological approaches to the treatment of obesity have not resulted yet in effective drugs. This review provides a panoramic of the multiple different approaches developed to obtain workable drugs. These approaches, however, rely in only four main lines of action: control of energy intake, mainly through modification of appetite;control of energy expenditure, essentially through the increase of thermogenesis;control of the availability of substrates to cells and tissues through hormonal and other metabolic factors controlling the fate of the available energy substrates; andcontrol of fat reserves through modulation of lipogenesis and lipolysis in white adipose tissue. A large proportion of current research is centred on neuropeptidic control of appetite, followed by the development of drugs controlling thermogenic mechanisms and analysis of the factors controlling adipocyte growth and fat storage. The adipocyte is also a fundamental source of metabolic signals, signals that can be intercepted, modulated and used to force the brain to adjust the mass of fat with the physiological means available. The large variety of different approaches used in the search for effective anti-obesity drugs show both the deep involvement of researchers on this field and the large amount of resources devoted to this problem by pharmaceutical companies. Future trends in anti-obesity drug research follow closely the approaches outlined; however, the increasing mass of information on the molecular basis of bodyweight control and obesity will in the end prevail in our search for effective and harmless anti-obesity drugs.
Collapse
Affiliation(s)
- José-Antonio Fernández-López
- Centre Especial de Recerca en Nutrició i Ciència dels Aliments, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|